research.vu.nl dissertation.pdf · ! The studies described in this thesis were carried out at the...

168

Transcript of research.vu.nl dissertation.pdf · ! The studies described in this thesis were carried out at the...

EXPLORING THE POTENTIAL ROLEOF THE PROTEIN, ALPHA-SYNUCLEIN, AS DIAGNOSTICTARGET FOR PARKINSON’SDISEASE.

NOUR K. MAJBOUR

 

The studies described in this thesis were carried out at the department of Anatomy

and Neurosciences of the VU University Medical Center, Amsterdam Neuroscience,

Amsterdam, the Netherlands, and College of Medicine and Health Sciences, United

Arab Emirates University, Al-Ain, UAE.

The study described in chapter 3 was partly funded by Parkinson Vereniging and

Amsterdam Neuroscience (to dr. van de Berg/prof dr. Berendse).

© 2019 by Nour Majbour, Amsterdam, the Netherlands.

All rights reserved. No part of this publication may be reproduced or transmitted in

any form or by any means without the prior written consent of the author.

 

ALPHA-SYNUCLEIN; THE MOST ATTRACTIVE MOLECULE IN PARKINSON’S DISEASE

EXPLORING THE POTENTIAL ROLE OF THE PROTEIN, ALPHA-

SYNUCLEIN, AS DIAGNOSTIC TARGET FOR PARKINSON’S DISEASE.

Nour K. Majbour                                                

 

VRIJE  UNIVERSITEIT    

EXPLORING  THE  POTENTIAL  ROLE  OF  THE  PROTEIN,  ALPHA-­‐SYNUCLEIN,    

AS  DIAGNOSTIC  TARGET  FOR  PARKINSON’S  DISEASE  

     

ACADEMISCH  PROEFSCHRIFT  ter  verkrijging  van  de  graad  Doctor  of  Philosophy  aan  

de  Vrije  Universiteit  Amsterdam,  op  gezag  van  de  rector  magnificus  

prof.dr.  V.  Subramaniam,  in  het  openbaar  te  verdedigen  

ten  overstaan  van  de  promotiecommissie  van  de  Faculteit  der  Geneeskunde  

op  maandag  11  maart  2019  om  15.45  uur  in  de  aula  van  de  universiteit,  

De  Boelelaan  1105          

door    

Nour  Khaled  Majbour  

geboren  te  Abu-­‐Dhabi,  Verenigde  Emiraten  

 

   

 

   

promotoren:         prof.dr.  O.M.A.  El-­‐Agnaf  prof.dr.  H.W.  Berendse  

copromotor:         dr.  W.D.J.  van  de  Berg                                                          

 

   beoordelingscommissie:     prof.  M.G.  Spillantini             prof.dr.ir.  C.E.  Teunissen                 prof.dr.  J.J.van  Hilten                     prof.dr.  R.M.A.  de  Bie             prof.dr.  W.M  van  der  Flier                    

 

7 GENERAL INTRODUCTION, AIM & OUTLINE OF THE THESIS

14 CHAPTER 1

Body Fluid Biomarkers in Parkinson’s Disease. Submitted to Nature Reviews Neurology

35 CHAPTER 2

Generation and Characterization of Novel Conformation-Specific Monoclonal Antibodies for Α-Synuclein Pathology. Neurobiology of Disease

75 CHAPTER 3

Oligomeric and Phosphorylated Alpha-Synuclein as Potential CSF Biomarkers for Parkinson’s Disease. Molecular Neurodegeneration

101 CHAPTER 4

Longitudinal Changes in CSF Alpha-Synuclein Species Reflect Parkinson's Disease Progression. Movement Disorders

116 CHAPTER 5

Increased Levels of CSF Total but Not Oligomeric or Phosphorylated Forms of Alpha-Synuclein in Patients Diagnosed with Probable Alzheimer's Disease. Scientific Reports

129 CHAPTER 6

CSF Oligomeric Alpha-Synuclein Levels as a Preclinical Marker of Parkinson’s Disease: A Study in LRRK2 Mutation Carriers and Sporadic Parkinson’s Disease Cases. Submitted to Neurology

149 CHAPTER 7

Summary & General Discussion

161 APPENDIX List of Publications

Word of Thanks About the Author

 

                   

   

To My Parents    

   

!!

GENERAL INTRODUCTION, AIM AND OUTLINE OF THE THESIS

  8  

 GENERAL INTRODUCTION,

AIM AND OUTLINE OF THE THESIS

 Although the first formal description of Parkinson disease (PD) by the English physician James Parkinson

in his essay on the “Shaking Palsy” was written more than 200 years ago, many aspects of the disease

remain to be elucidated. PD is a progressive, irreversible and age-related neurodegenerative disorder [1].

Of the many clinical symptoms that characterize PD, bradykinesia, muscle rigidity, resting tremor and

postural instability remain the cardinal ones [2]. In the recent years, we have come to appreciate that while

PD is a movement disorder, it also presents with a wide range of non-motor symptoms, such as cognitive

impairment, REM sleep behavior disorder, anxiety, depression and hyposmia [3]. Patients presenting with

bradykinesia in combination with muscle rigidity, resting tremor and/or postural instability are what

clinicians diagnose as PD. While severe autonomic involvement, cerebellar signs or early severe dementia

would rule out PD possibility. Clinical diagnosis also relies on progressive nature of the disease, a positive

effect of dopamine replacement therapy and a unilateral onset. [4]. Considering the progressive nature of

the disease and the overlapping of the clinical symptoms with other disorders such as essential tremor,

vascular parkinsonism, progressive supranuclear palsy (PSP), multiple system atrophy (MSA), diagnostic

accuracy of PD at the early stages yet to be improved [5, 6].

Neuropathologically, PD is mainly characterized by region-specific dopaminergic loss in the substantia

nigra pars compacta (SNpc) within the midbrain. Degeneration of the dopaminergic neurons results in

reduced levels of dopamine contributing to the motor dysfunction associated with PD [7]. In 1912, while

Friedrich Lewy was examining the brains of people who died with a clinical diagnosis of PD, he noticed

abnormal microscopic structures which are now known as Lewy bodies (LBs) [8] . LBs and Lewy neurites

(LNs) are the neuropathological hallmark of PD and dementia with Lewy bodies (DLB). LBs are

eosinophilic intracytoplasmic neuronal inclusions that are mainly composed of the protein alpha-synuclein

(α-syn) [8]. With that stated, Braak and Braak have also reflected on the fact that α-synuclein pathology

extends well beyond the substantia in their staging scheme [9].

α-Syn is a pre-synaptic neuronal protein and its aggregation and dysfunction is linked to a number of

neurodegenerative disorders named “synucleinopathies”. Actually, Since 1998 PD, dementia with Lewy

bodies (DLB) and multiple system atropgy (MSA) were frequently referred to as “synucleinopathies” [10].

The first antibodies against α-syn, produced in 1988, labeled both synapses and nuclei in the brain, leading

to the naming of synuclein [9, 11]. The precise function of this protein is not fully understood. However,

several lines of evidence through studies on different model organisms, suggest that α-syn is a major player

  9  

in vesicle trafficking, synaptic plasticity and neurotransmitter release [12]. α-Syn is subjected to several

post-translational modifications such as phosphorylation, oxidation, nitrosylation, truncation and

ubiquitination [13]. However, whether these post-translational modifications act to enhance or inhibit α-

syn neurotoxicity remains to be understood. . α-Syn is natively unfolded proteins, meaning that under

natural physiological conditions, it has a linear structure as random coils. However, numerous findings

suggest that α-syn aggregation plays a central role in the pathogenesis of synucleinopathies [14]. A better

understanding of the role of α-syn post-translational modifications may therefore help to elucidate the exact

role of α-syn in the pathogenesis of PD, paving the way for the development of new diagnostic and

therapeutic strategies for synucleinopathies. A number of findings have led to the speculation that the

aggregation of α-syn has a seminal role in PD. First, α-syn aggregates are the major component of LBs and

LNs. Second, several mutations in SNCA, the gene encoding α-syn protein, are associated with autosomal

dominantly inherited forms of PD [15]. Third, five missense α-syn mutations have been linked to rare forms

of early-onset familial PD [16-20], and they have been shown to increase α-syn aggregation in vitro.

furthermore, it has been also shown that mutant α-syn contain more β-sheet structure higher and causes

higher rates of α-syn self-oligomerization in vitro and amyloid fibrils formation compared to wild-type α-

syn [21, 22]. Based upon these observations, extensive efforts were put into the elucidation of pathogenic

mechanisms leading to α-syn polymerization and aggregation.

Despite major progress in our understanding of PD, we still lack effective disease-modifying therapies and

disease-specific biomarkers Treatments in most settings are symptomatic, and mainly focus on dopamine

replacement therapy. The clinical diagnostic criteria for PD have limited sensitivity and specificity,

particularly in the first years of the disease, mainly because PD is characterized by a striking heterogeneity

in clinical motor and non-motor features, rate of progression and susceptibility to the development of

adverse side effects. The struggle in detecting the disease at earlier stages, and the absence of reliable

methods to monitor disease progression or to assess response to treatments are other factors illustrating the

need for biomarkers. Many different approaches to the development of diagnostic and progression

biomarkers have emerged in recent years, with body fluid and brain imaging biomarkers at the forefront.

Biomarkers are objective measures of a disease state, disease progression and/or susceptibility to a disease.

Imaging techniques are increasingly employed to improve the accuracy of PD diagnosis. MRI scanning can

help to exclude secondary causes of parkinsonism [23], whereas single photon emission computed

tomography (SPECT) or Positron Emission Tomography (PET) can can facilitate the differential diagnosis

between PD and drug-induced, psychogenic and vascular parkinsonism or essential tremor. However, these

functional imaging techniques do not help to differentiate PD from other neurodegenerative parkinsonian

syndromes, such as MSA or PSP [24].

  10  

The great need for biomarkers that facilitate an early and reliable diagnosis of PD, preferably before the

actual development of clinical motor features, and the absolute necessity for monitoring disease progression

and response to treatment were the driving forces behind the work presented in the current thesis. The first

chapter of this thesis (Chapter 1) is a review of the progress made towards the development and validation

of PD biomarkers to improve disease detection and diagnosis. The original research conducted in the current

thesis primarily focuses on exploring the potential of the protein alpha-synuclein and its different forms as

biomarkers for PD in human CSF samples.

The key issues that we aimed to address in this thesis were:

1) Is it possible to overcome the limitations of pan antibodies to α-syn by generating conformation-

specific antibodies?

2) Can we develop robust methods to quantify the levels of α-syn forms in biological fluids from

patients and age-matched controls with optimal sensitivity and specificity?

3) Can the levels of CSF α-syn forms contribute to an early diagnosis of PD?

4) Can the levels of CSF α-syn forms reflect disease progression in PD?

5) Can the levels of CSF α-syn forms help identify individuals at risk of developing PD?

The development and thorough characterization of novel conformation-specific monoclonal antibodies for

α-syn is described in Chapter 2. Next, to measure α-syn in biofluids, we developed novel assays that can

overcome the limitations of conventional methods and serve as potential diagnostic tools for PD, as reported

in Chapter 3. Within the same chapter, our novel assays namely for t-, o- and pSer129-α-syn were deployed

in a cross-sectional analysis of human CSF samples from a Dutch cohort of PD patients and age-matched

healthy controls. In addition, we assessed the discriminating power of combining multiple CSF α-syn

species with classical AD biomarkers, and explored the correlation with clinical parameters. The next

logical question was whether α-syn species can serve as progression biomarkers for PD, and the answer

came from using the same assays to analyze samples from the longitudinal Deprenyl and Tocopherol

Antioxidative Therapy (DATATOP) for Parkinsonism study cohort, as detailed in Chapter 4. In short, the

DATATOP study was a multicenter, placebo-controlled clinical trial in patients with early-stage PD. All

patients were followed for an average of 2 years to an endpoint defined as the development of clinical

symptoms of sufficient severity to require dopamine replacement therapy. The study described in Chapter

5 was based upon a series of neuropathological findings of co-existence of α-syn and tau pathology in the

brains of patients with AD, PD and DLB, raising the question of whether α-syn species can improve the

diagnostic performance of classical AD biomarkers. This question was addressed in an Italian cohort of

CSF samples from AD patients who showed a CSF profile typical of AD at baseline as well as in cognitively

intact control subjects. Based upon the findings presented in the previous chapters, the next intriguing

  11  

question was whether α-syn would facilitate the diagnosis of PD at its prodromal stage or allow the

identification of high-risk individuals. To address this question we studied CSF levels of α-syn species in

symptomatic and asymptomatic leucine-rich repeat kinase 2 (LRRK2) mutation carriers, as well as in

sporadic PD patients and healthy control subjects, the results of which are described in Chapter 6. In the

last chapter, Chapter 7, we review and discuss the results of all individual chapters, portray the full picture

of the knowledge gained, and articulate how such knowledge can create a solid foundation for future studies.

  12  

References [1]   Sherwood,  "Parkinson,  J.  An  Essay  on  the  Shaking  Palsy,"  N.  &  and  Jones,  Eds.,  ed,  1817.  [2]   E.  R.  Dorsey  et  al.,  "Projected  number  of  people  with  Parkinson  disease  in  the  most  

populous  nations,  2005  through  2030,"  (in  eng),  Neurology,  vol.  68,  no.  5,  pp.  384-­‐6,  Jan  2007.  

[3]   A.  Schrag,  U.  F.  Siddiqui,  Z.  Anastasiou,  D.  Weintraub,  and  J.  M.  Schott,  "Clinical  variables  and  biomarkers  in  prediction  of  cognitive  impairment  in  patients  with  newly  diagnosed  Parkinson's  disease:  a  cohort  study,"  (in  eng),  Lancet  Neurol,  vol.  16,  no.  1,  pp.  66-­‐75,  Jan  2017.  

[4]   R.  B.  Postuma  et  al.,  "MDS  clinical  diagnostic  criteria  for  Parkinson's  disease,"  (in  eng),  Mov  Disord,  vol.  30,  no.  12,  pp.  1591-­‐601,  Oct  2015.  

[5]   J.  C.  Greenland,  C.  H.  Williams-­‐Gray,  and  R.  A.  Barker,  "The  clinical  heterogeneity  of  Parkinson's  disease  and  its  therapeutic  implications,"  (in  eng),  Eur  J  Neurosci,  Jul  2018.  

[6]   S.  M.  van  Rooden,  W.  J.  Heiser,  J.  N.  Kok,  D.  Verbaan,  J.  J.  van  Hilten,  and  J.  Marinus,  "The  identification  of  Parkinson's  disease  subtypes  using  cluster  analysis:  a  systematic  review,"  (in  eng),  Mov  Disord,  vol.  25,  no.  8,  pp.  969-­‐78,  Jun  2010.  

[7]   E.  Masliah  et  al.,  "Dopaminergic  loss  and  inclusion  body  formation  in  alpha-­‐synuclein  mice:  implications  for  neurodegenerative  disorders,"  (in  eng),  Science,  vol.  287,  no.  5456,  pp.  1265-­‐9,  Feb  2000.  

[8]   M.  G.  Spillantini,  R.  A.  Crowther,  R.  Jakes,  M.  Hasegawa,  and  M.  Goedert,  "alpha-­‐Synuclein  in  filamentous  inclusions  of  Lewy  bodies  from  Parkinson's  disease  and  dementia  with  lewy  bodies,"  (in  eng),  Proc  Natl  Acad  Sci  U  S  A,  vol.  95,  no.  11,  pp.  6469-­‐73,  May  26  1998.  

[9]   H.  Braak,  K.  Del  Tredici,  U.  Rüb,  R.  A.  de  Vos,  E.  N.  Jansen  Steur,  and  E.  Braak,  "Staging  of  brain  pathology  related  to  sporadic  Parkinson's  disease,"  (in  eng),  Neurobiol  Aging,  vol.  24,  no.  2,  pp.  197-­‐211,  2003  Mar-­‐Apr  2003.  

[10]   M.  Goedert  and  M.  G.  Spillantini,  "Lewy  body  diseases  and  multiple  system  atrophy  as  alpha-­‐synucleinopathies,"  (in  eng),  Mol  Psychiatry,  vol.  3,  no.  6,  pp.  462-­‐5,  Nov  1998.  

[11]   L.  Maroteaux,  J.  T.  Campanelli,  and  R.  H.  Scheller,  "Synuclein:  a  neuron-­‐specific  protein  localized  to  the  nucleus  and  presynaptic  nerve  terminal,"  (in  eng),  J  Neurosci,  vol.  8,  no.  8,  pp.  2804-­‐15,  Aug  1988.  

[12]   A.  Villar-­‐Piqué,  T.  Lopes  da  Fonseca,  and  T.  F.  Outeiro,  "Structure,  function  and  toxicity  of  alpha-­‐synuclein:  the  Bermuda  triangle  in  synucleinopathies,"  (in  eng),  J  Neurochem,  vol.  139  Suppl  1,  pp.  240-­‐255,  Oct  2016.  

[13]   A.  W.  Schmid,  B.  Fauvet,  M.  Moniatte,  and  H.  A.  Lashuel,  "Alpha-­‐synuclein  post-­‐translational  modifications  as  potential  biomarkers  for  Parkinson's  disease  and  other  synucleinopathies,"  (in  ENG),  Mol  Cell  Proteomics,  Aug  2013.  

[14]   M.  G.  Spillantini  and  M.  Goedert,  "Neurodegeneration  and  the  ordered  assembly  of  α-­‐synuclein,"  (in  eng),  Cell  Tissue  Res,  vol.  373,  no.  1,  pp.  137-­‐148,  Jul  2018.  

[15]   A.  B.  Singleton,  M.  J.  Farrer,  and  V.  Bonifati,  "The  genetics  of  Parkinson's  disease:  progress  and  therapeutic  implications,"  (in  eng),  Mov  Disord,  vol.  28,  no.  1,  pp.  14-­‐23,  Jan  2013.  

  13  

[16]   S.  Appel-­‐Cresswell  et  al.,  "Alpha-­‐synuclein  p.H50Q,  a  novel  pathogenic  mutation  for  Parkinson's  disease,"  (in  eng),  Mov  Disord,  vol.  28,  no.  6,  pp.  811-­‐3,  Jun  2013.  

[17]   R.  Krüger  et  al.,  "Ala30Pro  mutation  in  the  gene  encoding  alpha-­‐synuclein  in  Parkinson's  disease,"  (in  eng),  Nat  Genet,  vol.  18,  no.  2,  pp.  106-­‐8,  Feb  1998.  

[18]   M.  H.  Polymeropoulos  et  al.,  "Mutation  in  the  alpha-­‐synuclein  gene  identified  in  families  with  Parkinson's  disease,"  (in  eng),  Science,  vol.  276,  no.  5321,  pp.  2045-­‐7,  Jun  1997.  

[19]   C.  Proukakis  et  al.,  "A  novel  α-­‐synuclein  missense  mutation  in  Parkinson  disease,"  (in  eng),  Neurology,  vol.  80,  no.  11,  pp.  1062-­‐4,  Mar  2013.  

[20]   J.  J.  Zarranz  et  al.,  "The  new  mutation,  E46K,  of  alpha-­‐synuclein  causes  Parkinson  and  Lewy  body  dementia,"  (in  eng),  Ann  Neurol,  vol.  55,  no.  2,  pp.  164-­‐73,  Feb  2004.  

[21]   O.  M.  El-­‐Agnaf  et  al.,  "Aggregates  from  mutant  and  wild-­‐type  alpha-­‐synuclein  proteins  and  NAC  peptide  induce  apoptotic  cell  death  in  human  neuroblastoma  cells  by  formation  of  beta-­‐sheet  and  amyloid-­‐like  filaments,"  (in  eng),  FEBS  Lett,  vol.  440,  no.  1-­‐2,  pp.  71-­‐5,  Nov  1998.  

[22]   K.  A.  Conway,  J.  D.  Harper,  and  P.  T.  Lansbury,  "Fibrils  formed  in  vitro  from  alpha-­‐synuclein  and  two  mutant  forms  linked  to  Parkinson's  disease  are  typical  amyloid,"  (in  eng),  Biochemistry,  vol.  39,  no.  10,  pp.  2552-­‐63,  Mar  2000.  

[23]   A.  Schrag  et  al.,  "Differentiation  of  atypical  parkinsonian  syndromes  with  routine  MRI,"  (in  eng),  Neurology,  vol.  54,  no.  3,  pp.  697-­‐702,  Feb  2000.  

[24]   W.  E.  Weber  and  A.  M.  Vlaar,  "Role  of  DAT-­‐SPECT  in  diagnostic  work-­‐up  of  Parkinsonism,"  (in  eng),  Mov  Disord,  vol.  23,  no.  5,  pp.  774;  author  reply  774-­‐5,  Apr  2008.  

                               

  14    

!!

CHAPTER 1

Body Fluid Biomarkers in Parkinson’s Disease

  15  

CHAPTER 1 | Body Fluid Biomarkers in Parkinson’s Disease  Nour  K.  Majbour1,2,  Brit  Mollenhauer3,  Jing  Zhang4,  Takahiko  Tokuda5,  Wilma  D.  J.  van  de  Berg2,  

Henk  W.  Berendse6,  Omar  M.  A.  El-­‐Agnaf1*  1Neurological  Disorders  Research  Center,  Qatar  Biomedical  Research  Institute  (QBRI),  Hamad  Bin  Khalifa  University  (HBKU),  Education  City,  Qatar.  2Department  of  Anatomy  and  Neurosciences,  Neuroscience  Campus  Amsterdam,  VU  University  Medical  Centre,  Amsterdam,  The  Netherlands  3Paracelsus-­‐Elena-­‐Klinik,  Kassel,  Germany.  4University  of  Washington,  Seattle,  WA,  USA.  5Department   of   Neurology,   Research   Institute   for   Geriatrics,   Kyoto   Prefectural   University   of  Medicine,  Kyoto,  602-­‐0841  Japan  6Department   of   Neurology,   Neuroscience   Campus   Amsterdam,   VU   University  Medical   Centre,  Amsterdam,  The  Netherlands    Correspondence  to:  Prof.  Omar  M.A.  El-­‐Agnaf,    Neurological  Disorders  Center,  Qatar  Biomedical  Research  Institute,  Education  City,    Qatar  Foundation,    P.O.  Box  5825  Doha,    Qatar    Mobile:  (+974)  55  93  55  68  E-­‐mail:  [email protected]    Under  Review  by  Nature  Reviews  Neurology.  Oct  2018  

                             

  16  

Abstract  

Parkinson’s disease was first described over 200 years ago by James Parkinson, yet only in the

past two decades our understanding of the molecular underpinnings of this progressive disorder,

the neuropathological features and the genetic risk factors has increased at a revolutionary pace.

In spite of the current lack of objective biomarkers to diagnose Parkinson’s disease early in its

course or to serve as endpoints in clinical trials, tangible progress at identifying biomarkers has

been made over the past ten years. Growing attention has been paid to several promising biofluids

markers that could potentially contribute to disease diagnosis and monitoring of disease

progression or the effects of disease-modifying interventions. This Review highlights the progress

in the discovery and evolution of Parkinson’s disease diagnostic and progression biomarkers in

human body fluids, including the identification of at-risk individuals.

 

 

 

 

 

 

 

 

 

 

                         

  17  

I.     Introduction

Parkinson’s disease (PD) was described for the first time over two centuries ago. However, its

etiology remains an enigma 1. While a cure remains elusive, many promising therapeutic

interventions for PD have emerged over the past few years. Treatments in most settings rely heavily

on pharmacological interventions to alleviate symptoms, yet are ineffective in targeting underlying

pathological mechanisms. The clinical signs and symptoms of PD are not disease-specific, making

the conventional clinical criteria suboptimal. PD encompasses a wide range of motor and non-

motor symptoms with a large variation in disease-onset and progression among patients, making

the early diagnosis for PD very challenging 2,3. The gold standard remains the neuropathological

presence of aggregated α-synuclein and neuronal loss. The limitations surrounding the clinical

diagnosis of PD could be overcome by the implementation of disease-specific body fluid

biomarkers.

Another purpose for body fluid biomarkers is the identification of the disease in its earliest stages,

i.e. in the long premotor phase that characterizes PD 4. This premotor phase can serve as a window

for early therapeutic interventions aimed at slowing down or perhaps halt disease progression.

Therapeutic intervention is likely to hold the greatest impact during the early stages of the disease.

Currently, the absence of robust biomarkers is a major hurdle to conduct clinical trials of potential

disease-modifying therapies at the time when therapeutic intervention is likely to hold the greatest

impact. Ideally, a reliable biomarker or biomarker panel for PD should reflect the clinical disease

state as well as defined underlying mechanisms of PD neuropathology, be validated in

neuropathologically confirmed PD subjects, be able to establish an early diagnosis of PD and

distinguish it from other synucleinopathies. In addition, the biomarker (panel) should be cost-

effective, highly reproducible, minimally-invasive, and qualify as a surrogate endpoint in clinical

trials.

In PD, as in many other neurodegenerative disorders, the pathological accumulation of misfolded

proteins is the dominant feature underlying the disease. Alpha-synuclein (α-syn) is the main protein

implicated in the pathogenesis of PD, whereas, neuropathologically Lewy bodies (LBs) and Lewy

neurites (LNs) are the defining lesions of the disease 5. The conversion of monomeric α-syn into

soluble oligomers and protofibrils before maturating into insoluble amyloid fibrils, has attracted

researchers’ attention to identify diagnostic and therapeutic targets for PD and related disorders 6.

Of the many impediments facing the diagnosis of PD, the heterogeneity of the disease within the

PD spectrum and significant overlap with other neurodegenerative diseases are the main ones.

These diseases include dementia with Lewy bodies (DLB) and multiple system atrophy (MSA), the

so-called “synuclein aggregation disorders” as well as other neurodegenerative disorders, such as

  18  

Alzheimer’s disease (AD), where a significant portion of cases demonstrate Lewy bodies at autopsy

and/or vascular lesions 7. Neuroprotective strategies in PD aim to target among others α-syn

aggregation and therefore overlapping clinical features with non-synuclein aggregation disorders

are challenging in the implementation of a disease-specific therapy. In spite of the current lack of

objective biomarkers for PD, a tangible progress at identifying the main potential biomarkers has

been made over the past two decades. This will be addressed in the present review.

II. α-Synuclein; the most attractive target in PD pathology

It was 20 years ago, when two major findings brought α-syn to the forefront of PD aetiology and

pathogenesis. The first finding was done in 1997, when a missense mutation (A53T) identified in

SNCA, the gene encoding for α-syn, was linked to familial PD in families of Greek origin 8. It

wasn’t long before a second mutation that caused early-onset PD in a German pedigree was

discovered in 1998 in the same gene (A30P) 9. The second clue came from Spillantini and

colleagues, who showed that α-syn was the main component of LBs and LNs in idiopathic PD brain

tissue 5,10. Later on, duplication and triplication of SNCA were also linked to early-onset, rapid

progression cases of sporadic PD, imparting that elevated levels of wild-type α-syn would also put

individuals at greater risk of developing PD as well as worsening disease prognosis as a gene dosage

effect 11,12.

α-Syn is a 140 amino acid protein that is highly abundant in the brain, where it is primarily localized

in presynaptic terminals 13. It is also present in red blood cells, plasma, serum, cerebrospinal fluid

(CSF), saliva, skin nerves and many other peripheral tissues such as colon and submandilar glands 14,15. Although a great interest has emerged about the likely pathogenic significance of α-syn, its

normal function remains poorly understood. Several lines of evidence, through studies on different

model organisms, suggest that α-syn is a major player in vesicle trafficking, synaptic plasticity and

neurotransmitter release supported by its intracellular localization in the presynaptic terminals 16.

Until recently, not much was known about the role of α-syn in inflammation. This has now changed,

since many studies have highlighted the role of the protein in inducing innate and adaptive

immunity, while others described its implication in pro-inflammatory cascades in microglia or

mediator of astroglial inflammatory responses 17-19.

II.I Dear α-synuclein, what can you tell us about PD? Growing evidence favours the hypothesis that α-syn aggregation is central to the neurodegeneration

in synuclein aggregation disorders. Native α-syn exist as unfolded monomer, however, under

certain conditions, α-syn monomers undergo a conformational transition, in which soluble

monomers initially form oligomers, then gradually assemble into protofibrils and eventually form

  19  

the large insoluble amyloid fibrils resembling the filaments present in human LBs and LNs 20. The

factors that determine the accumulation and aggregation of α-syn are diverse, but summate to result

either in upregulation of the protein expression, impaired degradation or increased likelihood for

its oligomerization and aggregation 20. It was the discovery of α-syn in CSF and blood in early 2000

that shed light for the first time on α-syn as a putative biomarker for PD 14,21. Since then, many

research groups including ours have tried to quantify the levels of α-syn species in biological fluids,

most prominently total α-syn but with varying results.

II.II Total alpha-synuclein; good but not good enough Exploring α-syn as a diagnostic biomarker, cross-sectional analysis of CSF total-α-syn in most

studies have shown t-α-syn to be decreased either in early drug-naïve PD patients 22-24, or in patients

with moderate to advanced PD 25 compared to healthy controls or AD 26 (Steenoven et al.,

Movement Disorders 2018, in press). When testing plasma and saliva samples, which are easily

accessible fluids and thus more attractive targets for biomarker development, t-α-syn levels in these

fluids were not significantly different between PD patients and healthy subjects 25. Unfortunately,

the reduction in CSF t- α-syn, is not specific for PD, but has also been reported in other synuclein-

related disorders like DLB and MSA and with tremendous overlap also with non-synuclein

disorders. In most studies exploring whether t-α-syn could reflect disease severity, an inverse

correlation with cognitive impairment was observed.

The role that t-α-syn could play as a progression biomarker does not seem to be sufficient either.

In longitudinal cohort studies, CSF levels of t-α-syn remained relatively stable over a follow-up

duration of either 24 months in the PPMI study or up to 4 years in the Norwegian ParkWest study 27,28. In other longitudinal studies CSF t-α-syn either showed a longitudinal increase over the early

course of the disease 29,30, or a significant decrease with PD progression 31. Two studies found

baseline CSF t-α-syn to predict cognitive decline in PD over the course of the disease 31,32. Although

most of the t-α-syn biomarker studies were performed in well-controlled cohorts, there are a

number of factors that may explain the inconsistencies in the above-described observations. These

include pre-analytical factors concerning the collection and the processing of the samples, the assay

platforms and the heterogeneity of patient and controls groups. In this context, the immediate

question is, could we do better when focussing on other species of α-syn?

II.III Oligomeric alpha-synuclein; know the enemy, make it your ally The debate about the most relevant α-syn inclusions to disease pathology has been going on for

some time. While growing evidence tips the scale in favour of soluble α-syn oligomers (o-α-syn)

being directly associated with neurodegeneration, the nature of the exact neurotoxic species

  20  

remains a point of controversy. The work of many researchers reflected strongly on the

neurotoxicity of α-syn oligomers 16,33,34.

The observation of a significant increase of CSF o-α-syn in PD patients compared to controls by

our group and others 35 brought o-α-syn to the forefront as potential biomarker for PD. When Park

and coworkers quantified α-syn oligomers in both CSF and plasma from drug naïve patients using

ELISA platform 36, CSF o-α-syn levels were elevated in PD compared to control subjects, whereas,

the difference in serum o-α-syn levels did not reach statistical significance. Another study by Aasly

et al. using an an improved version of previously described ELISA 35 explored the potential of t-

and o-α-syn in a LRRK2 cohort 37. Interestingly, o-α-syn was significantly higher in LRRK2 carriers

as well as in sporadic PD subjects compared to healthy controls. Similarly, Parnetti et al., found o-

α-syn to be significantly elevated in PD compared to neurological controls 38. An assessement of

the levels of CSF o-α-syn in a cohort of patients with the most common forms of dementia, i.e.

PDD, DLB and AD, revealed that the levels of o-α-syn were significantly increased in

synucleinopathy dementias rather than in tauopathy dementias 39. While this study didn’t fully

answer whether o-α-syn can serve as potential biomarker for dementia, clues were given of o-α-

syn being more intimately connected to synucleinopathies than tauopathies.

The considerable overlap noted among the diagnostic groups in these studies was revealing. It made

clear that while the method was successful, the assays had limitations. The studies were flawed by

the absence of a calibrator for the oligomeric ELISA, which roadblocked us from understanding

the levels of o-α-syn in terms of the actual concentrations and their relationship to t-α-syn

concentration. Another limitation was the lack of oligomeric-specific antibodies which could

overcome the hurdles with the previous assays. The first oligomeric-specific ELISA using

conformation-specific monoclonal antibodies for PD pathology was described in 2016 22. In this

study by El-Agnaf and coworkers levels of CSF o-α-syn were analysed using conformation-specific

antibodies built-in ELISA, as well as a thoroughly characterized calibrator of α-syn oligomers. The

CSF levels of o-α-syn using the new ELISA were indeed higher in PD, similar to what was reported

before, but the overlap was minimized between the two groups. More recently, in a cross-sectional

analysis of CSF o-α-syn levels in a cohort of PD, DLB, AD and controls with subjective cognitive

decline, o-α-syn was markedly elevated in both PD and DLB patients compared to AD and controls

subjects (Steenoven et al., Movement Disorders 2018, in press). Among the very few studies

assessing longitudinal changes in CSF biomarkers over the course of PD, only one covered CSF o-

α-syn. In the longitudinal DATATOP cohort, a longitudinal increase in o-α-syn over the two-year

follow up period, and a strong positive correlation between the changes in o- and t-α-syn were

observed, unravelling the dynamic pattern of o-α-syn over the early course of PD 29.

  21  

In recent years the seeding propensity of α-syn in biological fluids has gained increasing interest.

When CSF samples of PD patients and control subjects with other neurological disorders where

screened using protein misfolding cyclic amplification (PMCA) technology, the specificity for PD

reached 96.9% with a sensitivity of 88.5%. While PMCA technology is quite promising, many

questions such as monitoring disease progression or predicting disease in the pre-motor phase

remain to be answered. Inspired by PMCA, RT-QuIC was developed by Caughey for detecting

different types of prions 40. RT-QuIC captures the same principle as PMCA, but runs differently to

save time, cost and effort. It wasn’t long before Parkkinen and coworkers tailored RT-QuIC to

detect o-α-syn in CSF of patients with DLB with 92% and 95% sensitivity and specificity,

respectively 41. With the goal of improving RT-QuIC practicality, Caughey and coworkers reduced

the assay time and gave it a quantitative feature 42. The analysis of CSF samples from PD and DLB

cases compared to non synucleinopathies controls, revealed a 93% diagnostic sensitivity and 100%

specificity. While the potential of RT-QuIC seems remarkable, many challenges of the assay still

have to be overcome; RT-QuIC can only detect forms of α-syn that can be seeded, while other

forms of α-syn that can not be seeded are also related to disease pathogenesis. Other issues include

the following questions: Would different strains of α-syn exempt the same seeding kinetics? Could

RT-QuIC be used in biofluids collected by less invasive methods, such as plasma, serum or saliva?

The most important questions still remain: Would any of the oligomeric-specific assays provide an

early diagnosis for PD prior to the manifestation of the clinical symptoms? And could they help

us monitor disease progression?

II.IV Phosphorylated S129 alpha-synuclein; harmful or beneficial? An increasing number of studies reported that α-syn is subjected to several post translational

modifications (PTMs), such as phosphorylation, ubiquitination, sumoylation, nitration or c-

terminal truncation, changing the protein conformation and/or function 43,44. Moreover, these

studies suggested that PTMs may play a critical role in regulating α-syn aggregation and toxicity

in vivo. Considering that only a small fraction of α-syn (~ 4%) is phosphorylated at residue Ser 129

under physiological conditions in vivo, whereas almost 90% of α-syn in LBs is phosphorylated at

the same residue, the significant attention that was devoted to pS129-α-syn is easily explained.

While it is clear that pS129-α-syn can alter its characteristics in terms of aggregation and/or

neurotoxicity, whether phosphorylation promotes or protects against PD is less clear 43. Regardless

of how future research will answer this question, pS129-α-syn is a potential biomarker for PD.

Unfortunately, quantitative assessment of pS129-α-syn in biological fluids turned out to be more

challenging than initially anticipated, due to the scarcity of pS129-α-syn specific antibodies, the

abundance of phosphatases in biofluids and the questioned stability of pS129-α-syn over long

  22  

storage of the samples. As a result, only few laboratories were able to quantify pS129-α-syn in CSF

or blood.

In 2012, Zhang and co-workers first used reported a sensitive and specific assay for pS129-α-syn

to measure CSF pS129-α-syn in two sets of PD cohorts, a discovery cohort and a validation cohort 45. While pS129-α-syn discriminated PD patients from healthy subjects in the discovery set, this

could not be reproduced in the validation set. However, in both cohorts pS129-α-syn levels were

significantly higher in PD compared to MSA and PSP. In addition, a weak correlation between CSF

pS129-α-syn levels and PD severity was reported. The same assay was subsequently used to

examine the longitudinal changes of pS129-α-syn in the DATATOP cohort. Compared to baseline,

an insignificant increase in pS129-α-syn was observed. Close examination of the correlation

between CSF pS129-α-syn and disease severity in subjects from DATATOP, a large cross-sectional

cohort, and a cohort of LRRK2 mutation carriers after stratification by PD stage, revealed a

negative-to-positive transition over the different disease stages. Following a U-shaped curve, the

inverse correlation between CSF pS129-α-syn and disease severity at early stages changes to a

positive correlation at later stages. This knowledge can help us determine the optimal way in which

pS129-α-syn can be used as a biomarker for PD 46. Using a pS129-α-syn specific ELISA, first in a

cross-sectional cohort and thereafter in a longitudinal cohort, we found that CSF pS129-a-syn levels

are significantly higher in PD compared to HC, emphasizing the potential of pS129-α-syn as a

diagnostic marker for PD. Over a period of two years of disease progression, a decrease of pS129-

α-syn levels was noted at follow-up compared to baseline. The discrepancies between our findings

and those of Stewart et al. can be explained by differences in selection criteria of the patients from

DATATOP in each study. Stewart et al., included unmediated PD patients from the placebo group

that were followed longer than 6 months (n=95), whereas in Majbour et al., 121 patients with

definite PD (90%-100% confidence based on the investigators’ report (n=121) were selected. Other

factors that may have contributed to these discrepancies include the protocols used for processing

the samples, the methods used for quantification, and the antibodies used. More recently, CSF

pS129-α-syn were unchanged when assessed in PD and DLB compared to AD and patients with

subjective cognitive decline (Steenoven et al., Movement Disorders 2018, in press).

II.V Combining multiple species of α-syn; team work always pays off Combining multiple species of α-syn along with other biomarkers has been the most promising

approach for diagnostic purposes in PD thus far. Combining o- and t-α-syn in an o-/t-α-syn ratio, a

sensitivity of 89.3% and specificity of 90.6%, with an AUC of 0.948 was achieved for a diagnosis

of PD 35. In a study exploring the diagnostic value of several biological markers, t-α-syn, tau, Aβ42

and total protein in CSF and serum samples from three cross-sectional cohorts of patients with

  23  

different synucleinopathies (PD, MSA or DLB), AD, parkinsonism or other neurological disorder,

CSF t-α-syn and tau along with the age of participants, provided the best discriminative model for

distinguishing patients with synucleinopathies from patients with AD or other neurological

disorders 47.

In parallel with the findings above, Wang et al. showed how a combination of pS129- and t-α-syn

could distinguish PD from MSA and PSP 45. In another study, combining a wider range of

biomarkers, namely t- and o-α-syn, Aβ42 and tau, provided the best diagnostic accuracy for

differentiating between PD patients and patients with other neurological disorders 48. Using our

specific ELISA assays, we found that both pS129-/t-α-syn and o-/t-α-syn ratios improved the

discrimination between PD and healthy control subjects 22. The best predictive model for

discriminating PD could be generated by combining these ratios with p-tau levels in a logistic

regression analysis. Interestingly, also in the DATATOP cohort, in which a correlation between

single species of α-syn and PD severity was absent, combining o- and t-α-syn resulted in a fair

negative correlation with motor dysfunction 29. Remarkably, when PD patients where sub-grouped

based on their clinical phenotype to tremor dominant, postural instability and gait difficulty

dominant, or intermediate, the correlation between o-/t-α-syn ratio and the motor impairment was

stronger within the tremor dominant group and absent in the postural instability and gait difficulty

group, thus for the first time linking CSF biomarkers with clinical phenotype 29. Such extensive

cohort analyses strongly emphasize the significance of combining the measurement of biological

markers, perhaps even from different biofluids, along with patients’ characteristics (e.g. age) in

developing optimal therapeutic approaches as well as directing clinical trials towards the right

population.

III. Uric Acid; the lower the levels, the higher the risk

There is no doubt that oxidative stress plays a critical role in the neurodegenerative process of PD,

and it’s no secret that uric acid is among the antioxidants meant to suppress oxidative stress and

thus protects against cell death 49. Uric acid is therefore another potential biomarker for PD that has

received little attention so far, yet may reflect a mechanism underlying PD pathology. Data

highlighting the likely usefulness of uric acid as a biomarker for PD mainly emanated from

epidemiological and genetic studies, and a recent clinical trial 50,51. While studies assessing the

levels of uric acid in blood are limited in number, even fewer studies were aimed at CSF levels.

Most of these studies demonstrated reduced levels of uric acid in patients with PD compared to

control subjects, hence proposing that reduced levels of uric acid put individuals at a higher risk of

developing PD and vice versa.

  24  

IV. Lysosomal Enzymes; tricky to measure

The accumulating evidence about the link between lysosomal dysfunction and an increased risk of

PD 52, propelled the efforts of many researchers to explore the potential of CSF lysosomal enzymes

as biomarkers for PD. However, only few studies were successful. In a small cohort of PD and

neurological controls, the levels of CSF GCase were reduced in PD patients compared to

individuals suffering from other neurological conditions 53. When tested in a larger cohort, CSF

GCase levels better discriminated PD from control subjects when combined with CSF o-/t-α-syn

ratio with a sensitivity of 82% and a specificity of 71%, which once more emphasizes the

importance of combining multiple biomarkers to improve diagnostic accuracy of PD 54. Substantial

overlap between PD and controls as well as the many factors interfering with the quantification of

CSF lysosomal enzymes are among the reasons why lysosomal enzymes have a long way to go

before they can be used in clinical practice as surrogate biomarkers for PD 52,55.

V. Neurofilament Light Chain; a differential biomarker? Increased CSF levels of Neurofilament Light Chain (NF-L) are a strong marker of axonal injury.

Most studies have focused on the discriminative power of NF-L to differentiate PD patients from

other forms of parkinsonism, in particular MSA and PSP patients.

In a study by Holmberg and co-workers, involving a cohort of carefully characterized patients with

PD, MSA or PSP, CSF levels of NF-L were significantly higher in MSA and PSP patients compared

to PD. The discriminative analysis showed a sensitivity of 78% and a specificity of 80%, with only

7 PD patients out of 210 being false-positives 56. Similarly, others have observed a significantly

higher level of NF-L in MSA patients compared to PD with a specificity of 90% and a sensitivity

of 83% with AUC of 0.92 57. Subsequently, Holmberg and colleagues reported similar findings,

this time including another atypical parkinsonion disorder (corticobasal degeneration) as well as a

control group of healthy subjects 58. Although CSF NF-L levels segregated atypical parkinsonion

disorders from PD, they failed to discriminate PD patients from healthy controls which precludes

NF-L CSF levels from being an optimal stand-alone biomarker for PD. This finding was later

confirmed by Hall et al., reporting significantly higher CSF NF-L levels in patients with atypical

parkinsonion disorders (MSA, PSP or CBD) compared to PD and healthy subjects 59. In the same

study, positive correlations were found between CSF NF-L levels and motor function in PD and

PSP, and between CSF-NF-L and cognitive decline in AD. A positive correlation with age was also

observed in all diagnostic groups 59.

To understand the full potential of NF-L as a biomarker for PD, the same research group went a

step further to assess the levels of NF-L in blood 60. Interestingly, CSF levels of NF-L positively

and strongly correlated with NF-L levels in blood in all groups from the three independent cohorts.

  25  

The blood levels of NF-L also distinguished PD from other atypical parkinsonion disorders.

Hansson et al. not only validated the importance of NF-L as a potential biomarker for PD but also

the accessibility of this biomarker in convenient biofluids like blood 60.

To summarize, CSF and serum NF-L is an attractive biomarker to distinguish between PD and

atypical parkinsonian disorders. It is easily accessible and its levels correlate with disease severity,

but NF-L can not be used to differentiate PD from DLB or healthy subjects.

VI. Inflammatory Biomarkers; nonspecific, yet appealing Inflammation as a pathogenic factor in neurodegeneration has become a hot topic in the past few

years. Converging evidence supports the involvement of central and peripheral inflammation in the

pathogenesis of PD. Some researchers favour the hypothesis that inflammation is a causative factor,

while others believe it is merely a by-product of neurodegeneration. In both cases, inflammatory

markers might hold a promise as candidate biomarkers for PD diagnosis or monitoring of disease

progression. Many researchers have addressed this issue by measuring inflammatory markers in

serum and CSF samples of PD patients and healthy controls. The results of a recent study 61 suggests

that inflammatory markers are relatively stable and thus can be reliably measured. In spite of that

observation, conflicting results have been obtained. Most studies agree that serum and CSF IL-2,

IL-6, IL-10, IL-1β, tumour necrosis factor, C-reactive protein (CRP), neutrophil gelatinase-

associated lipocalin and IFNγ occupy the highest ranks among potential inflammatory markers 61,62.

In most studies, the levels of these markers were significantly higher in PD patients compared to

healthy control subjects. However, none of these markers optimally discriminated PD patients from

controls as a single marker, most probably reflecting heterogeneity of the disease. When exploring

the association between CSF inflammatory markers and non-motor symptoms in PD patients,

Hansson and co-workers observed that CRP levels were significantly higher in PD patients with

dementia (PDD) compared to non-demented PD patients or healthy controls, whereas all other

inflammatory biomarkers showed no significant difference 63. Nevertheless, the degree of

neuroinflammation in the PD group significantly correlated with more severe depression, fatigue,

and cognitive impairment 63.

In a study by Maetzler et al., involving 142 LRRK2-positive PD patients, levels of inflammatory

biomarkers, including interleukin 8 (IL-8), monocyte chemotactic protein 1 (MCP-1) and

macrophage inflammatory protein 1-b (MIP-1-b), discriminated between patients classified as

having a diffuse/malignant phenotype compared to patients classified as intermediate or mainly

pure motor 64.

In 2018, Karpenko et al., investigated the potential of inflammatory markers in CSF and serum of

PD patients compared to healthy subjects 65. In CSF, no significant differences in inflammatory

  26  

markers were noted between PD and control groups. In serum, however, significantly lower IL-

1RA and higher IL-Iβ and IL-6 were observed in the PD group. CSF IL-6 inversely correlated with

disease duration and severity, measured as Hoehn-Yahr stage and UPDRS II scores, while TNFα

positivity correlated with disease progression. Serum IL-6 positively correlated with both PD

severity and rate of progression, and inversely with disease duration. Exploring the relationship

between inflammatory markers and non-motor symptoms of PD, increased levels of IL-10 in serum

correlated with increased anxiety and depression, whereas serum TNFα levels were lower in PD

patients with mild cognitive impairment compared to healthy controls 65.

VII. AD-associated biomarkers; the question of complexity arises In recent years, it has been increasingly recognized that PD is a motor disorder with non-motor

features that are equally important 4,66. Mild cognitive impairment occurs in about 20-50% of

patients with PD, whereas the prevalence of dementia rises up to 75% in patients who have had PD

for more than 10 years 67. From a neuropathological standpoint, co-occurrence of PD and AD

pathologies has repeatedly been reported, suggesting that there may be an interaction between PD

and AD pathologies, i.e. α-syn, Aβ and tau proteins, in human brain and in animal models 68,69.

The main AD protein biomarkers that have been widely investigated in PD pathology are tau,

phosphorylated tau, Aβ40 and Aβ42, mostly focusing on the link between Aβ levels and cognitive

decline 70. In a study performed by Compta et al., the levels of tau, p-tau and Aβ were assessed in

CSF from PD and PDD patients, and in healthy subjects 71. CSF tau and p-tau were significantly

elevated in PDD patients compared to non-demented patients or healthy subjects. In addition, CSF

tau and p-tau levels were inversely correlated with memory impairment. Aβ42 on the other hand,

was lowest in PDD patients and positively correlated with phonetic fluency. It is noteworthy

mentioning that the small size of the subgroups undermines the power of these findings. Most cross-

sectional studies reported reduced levels of Aβ42 in PD patients compared to healthy controls 71,72

, elevated CSF tau and p-tau levels in PDD patients 71 while unchanged in non-demented PD

patients compared to healthy controls 22,38,72. The discrepancies between these findings may be

explained by differences in the clinical profile of the included patients and whether they were

medicated or drug-naïve. With respect to longitudinal changes, over a follow-up period of one to

two years in most studies, CSF Aβ42 levels showed a decreasing trend with lower baseline levels

being associated with higher risk of developing cognitive impairment, whereas tau and p-tau levels

remained relatively stable and showed no correlation with cognitive decline 29,73,74.

  27  

VIII. Conclusions and Future Perspectives; Then, Now and Future Considering the developments in the field of body fluid biomarkers in PD over the past two

decades, it is clear that it will be a daunting feat to reach a reliable diagnosis for PD based on a

simple test in a blood or CSF sample. The main challenges that delay the discovery of reliable PD

biomarkers are: the complex nature of the disease, the overlap with other neurodegenerative

diseases, the heterogeneity of the clinical phenotypes, and the many missing pieces of the puzzle

underpinning the disease etiology. In spite of these obstacles, as described in this review, the field

of biomarkers for PD has rapidly advanced on several fronts over the past ten years. There have

been concentrated and joint efforts by researchers and funding agencies for Parkinson’s Research

to spur the identification of reproducible biomarkers for PD. Since the ideal (panel) of biomarker(s)

has not yet been identified, the pursuit is still going on.

The biggest achievement in the last years was the recruitment of patient cohorts with the

longitudinal collection of biological fluids, such as the Parkinson’s Progression Marker Initiative

(www.ppmi-info.org). In addition, we have witnessed other development that are worthy of

recognition: tools and technologies have been developed to more accurately assess biomarkers 22,41,75,76, the spectrum of biological samples is widening (CSF, plasma, serum, saliva, skin biopsies,

etc.), the array of potential biomarkers is expanding (oligomeric α-syn, phosphorylated α-syn,

lysosomes and inflammatory biomarkers), the value of using a panel of biomarkers rather than a

single marker has become clearer, and the interest in longitudinal studies is deepening. We have

also come to appreciate the importance of pre-analytical factors; sample collection, processing,

thawing cycles, selection of tips and tubes for handling the samples, haemoglobin contamination

and storage conditions 77. These factors urged research centers to put collective efforts into

developing standardized protocols and methodologies for sample processing 78. As reviewed here,

it has become clear that a single biomarker can not stand alone for PD diagnosis or progression.

Instead, a panel of at least 5 markers is probably required to cover the clinical heterogeneity of PD.

From a diagnostic perspective, the main answers that research has delivered are 1) reduced serum

uric acid levels and elevated serum IL_6 and IL-10 levels can aid in the identification of individuals

at risk of PD, 2) CSF and serum NF-L levels are the best candidate biomarker to distinghuish PD

patients from patients with other parkinsonian disorders, 3) a combination of α-syn species, mainly

t- and o-a-syn can distinguish PD patients from subjects without a synucleinopathy (either healthy

controls or patients with other neurological disorders), 4) Aβ42 can differentiate between PDD and

PD patients. With respect to monitoring of disease progression, the levels of only some biomarkers

were associated with worsening PD motor and non-motor symptoms. Levels of CSF t-a-syn, CSF

Aβ42 and serum inflammatory markers correlated with cognitive decline, the strength and direction

  28  

of the correlation varying from one marker to another. While o-a-syn appears to reflect PD-related

motor deterioration, the ratio of o-/t-a-syn better reflects longitudinal disease progression and PD

clinical phenotypes. Taken together, there is increasing evidence that a panel of potentially

informative biomarkers for PD is coming together, which should at least include CSF t-a-syn, CSF

o-a-syn, CSF NF-L, Aβ42 and serum inflammatory markers (Figure 1).

In order to move forward, major questions remain to be answered by future studies; what is the

optimal method to determine cut-off points for chemical biomarkers? What does it take to

implement body fluid biomarkers into routine clinical care? Can we use blood biomarkers instead  

of CSF biomarkers? Although the quest to find an ideal biomarker for PD has certainly not ended,

much ground has been covered and we are getting closer to the ultimate goal. We should therefore

sustain the momentum of the progress made towards the development of the ideal biomarker panel

for PD.

 

   

 

 

  29  

 

 

         

             

Figure.(1(

Uric%AcidInflammatory%markers

Neurofilament6light%chain

⍺6Synuclein%Forms

ADPD

PDD

DLB

HC

AD%biomarkers

At%risk

Atypical%Parkinsonism

DLBPD

PDD

PDD

AD

HC

PD

ADPD

PDD

DLB

Atypical-Parkinsonism

HC

CSF Blood

ADPD

At%riskPDD

DLB

Atypical%Parkinsonism

HC

Figure'1.(Proposed(Panel(of(Biomarkers(for(Parkinson’s(Disease(Differential(Diagnosis. The(chart(highlights(the(value(of(using(a(panel(of(biomarkers(to( improve(the(differential(diagnosis(of(PD.(Levels(of(serum(uric(acid(and(inflammatory(markers,(mainly(IL_6(and(ILD10(can(aid(identify(the(individuals(at(risk(of(PD.(CSF(and(serum(NFDL(can(distinguish(PD(patients(from(patients(with(other(parkinsonion(disorders((atypical( parkinsonism).( A( combination( of( αDsyn( species,( can( distinguish( PD( patients( from( HC,( DLB,( AD(subjects.(AD(biomarkers,(namely(CSF(Aβ42(can(differentiate(between(PDD(from(PD(patients. PD:(Parkinson’s(disease,(DLB:(Dementia(with(Lewy(bodies,(AD:(Alzheimer’s’(disease,(PDD:(Parkinson’s(disease(with(dementia,(HC:(healthy(controls

  30  

References  1   Sherwood.        (eds  Neely  &  &  Jones)  (1817).  2   Postuma,  R.  B.  et  al.  MDS  clinical  diagnostic  criteria  for  Parkinson's  disease.  Mov  Disord  

30,  1591-­‐1601,  doi:10.1002/mds.26424  (2015).  3   Marsili,  L.,  Rizzo,  G.  &  Colosimo,  C.  Diagnostic  Criteria  for  Parkinson's  Disease:  From  

James  Parkinson  to  the  Concept  of  Prodromal  Disease.  Front  Neurol  9,  156,  doi:10.3389/fneur.2018.00156  (2018).  

4   Schrag,  A.,  Horsfall,  L.,  Walters,  K.,  Noyce,  A.  &  Petersen,  I.  Prediagnostic  presentations  of  Parkinson's  disease  in  primary  care:  a  case-­‐control  study.  Lancet  Neurol  14,  57-­‐64,  doi:10.1016/S1474-­‐4422(14)70287-­‐X  (2015).  

5   Goedert,  M.  Alpha-­‐synuclein  and  neurodegenerative  diseases.  Nat  Rev  Neurosci  2,  492-­‐501,  doi:10.1038/35081564  (2001).  

6   Conway,  K.  A.,  Harper,  J.  D.  &  Lansbury,  P.  T.  Fibrils  formed  in  vitro  from  alpha-­‐synuclein  and  two  mutant  forms  linked  to  Parkinson's  disease  are  typical  amyloid.  Biochemistry  39,  2552-­‐2563  (2000).  

7   Lee,  V.  M.,  Giasson,  B.  I.  &  Trojanowski,  J.  Q.  More  than  just  two  peas  in  a  pod:  common  amyloidogenic  properties  of  tau  and  alpha-­‐synuclein  in  neurodegenerative  diseases.  Trends  Neurosci  27,  129-­‐134,  doi:10.1016/j.tins.2004.01.007  (2004).  

8   Spira,  P.  J.,  Sharpe,  D.  M.,  Halliday,  G.,  Cavanagh,  J.  &  Nicholson,  G.  A.  Clinical  and  pathological  features  of  a  Parkinsonian  syndrome  in  a  family  with  an  Ala53Thr  alpha-­‐synuclein  mutation.  Ann  Neurol  49,  313-­‐319  (2001).  

9   Gasser,  T.  et  al.  A  susceptibility  locus  for  Parkinson's  disease  maps  to  chromosome  2p13.  Nat  Genet  18,  262-­‐265,  doi:10.1038/ng0398-­‐262  (1998).  

10   Spillantini,  M.  G.  et  al.  Alpha-­‐synuclein  in  Lewy  bodies.  Nature  388,  839-­‐840,  doi:10.1038/42166  (1997).  

11   Singleton,  A.  B.  et  al.  alpha-­‐Synuclein  locus  triplication  causes  Parkinson's  disease.  Science  302,  841,  doi:10.1126/science.1090278  (2003).  

12   Chartier-­‐Harlin,  M.  C.  et  al.  Alpha-­‐synuclein  locus  duplication  as  a  cause  of  familial  Parkinson's  disease.  Lancet  364,  1167-­‐1169,  doi:10.1016/S0140-­‐6736(04)17103-­‐1  (2004).  

13   Maroteaux,  L.,  Campanelli,  J.  T.  &  Scheller,  R.  H.  Synuclein:  a  neuron-­‐specific  protein  localized  to  the  nucleus  and  presynaptic  nerve  terminal.  J  Neurosci  8,  2804-­‐2815  (1988).  

14   Borghi,  R.  et  al.  Full  length  alpha-­‐synuclein  is  present  in  cerebrospinal  fluid  from  Parkinson's  disease  and  normal  subjects.  Neurosci  Lett  287,  65-­‐67  (2000).  

15   Visanji,  N.  P.  et  al.  The  Systemic  Synuclein  Sampling  Study:  toward  a  biomarker  for  Parkinson's  disease.  Biomark  Med  11,  359-­‐368,  doi:10.2217/bmm-­‐2016-­‐0366  (2017).  

16   Villar-­‐Piqué,  A.,  Lopes  da  Fonseca,  T.  &  Outeiro,  T.  F.  Structure,  function  and  toxicity  of  alpha-­‐synuclein:  the  Bermuda  triangle  in  synucleinopathies.  J  Neurochem  139  Suppl  1,  240-­‐255,  doi:10.1111/jnc.13249  (2016).  

17   Kannarkat,  G.  T.,  Boss,  J.  M.  &  Tansey,  M.  G.  The  role  of  innate  and  adaptive  immunity  in  Parkinson's  disease.  J  Parkinsons  Dis  3,  493-­‐514,  doi:10.3233/JPD-­‐130250  (2013).  

18   Lee,  H.  J.  et  al.  Direct  transfer  of  alpha-­‐synuclein  from  neuron  to  astroglia  causes  inflammatory  responses  in  synucleinopathies.  J  Biol  Chem  285,  9262-­‐9272,  doi:10.1074/jbc.M109.081125  (2010).  

  31  

19   Zhang,  W.  et  al.  Aggregated  alpha-­‐synuclein  activates  microglia:  a  process  leading  to  disease  progression  in  Parkinson's  disease.  FASEB  J  19,  533-­‐542,  doi:10.1096/fj.04-­‐2751com  (2005).  

20   Spillantini,  M.  G.  &  Goedert,  M.  Neurodegeneration  and  the  ordered  assembly  of  α-­‐synuclein.  Cell  Tissue  Res,  doi:10.1007/s00441-­‐017-­‐2706-­‐9  (2017).  

21   El-­‐Agnaf,  O.  M.  et  al.  Alpha-­‐synuclein  implicated  in  Parkinson's  disease  is  present  in  extracellular  biological  fluids,  including  human  plasma.  FASEB  J  17,  1945-­‐1947,  doi:10.1096/fj.03-­‐0098fje  (2003).  

22   Majbour,  N.  K.  et  al.  Oligomeric  and  phosphorylated  alpha-­‐synuclein  as  potential  CSF  biomarkers  for  Parkinson's  disease.  Mol  Neurodegener  11,  7,  doi:10.1186/s13024-­‐016-­‐0072-­‐9  (2016).  

23   Kang,  J.  H.  Cerebrospinal  Fluid  Amyloid  β1-­‐42,  Tau,  and  Alpha-­‐Synuclein  Predict  the  Heterogeneous  Progression  of  Cognitive  Dysfunction  in  Parkinson's  Disease.  J  Mov  Disord  9,  89-­‐96,  doi:10.14802/jmd.16017  (2016).  

24   Mollenhauer,  B.  et  al.  Total  CSF  α-­‐synuclein  is  lower  in  de  novo  Parkinson  patients  than  in  healthy  subjects.  Neurosci  Lett  532,  44-­‐48,  doi:10.1016/j.neulet.2012.11.004  (2013).  

25   Goldman,  J.  G.  et  al.  Cerebrospinal  fluid,  plasma,  and  saliva  in  the  BioFIND  study:  Relationships  among  biomarkers  and  Parkinson's  disease  Features.  Mov  Disord  33,  282-­‐288,  doi:10.1002/mds.27232  (2018).  

26   Eusebi,  P.  et  al.  Diagnostic  utility  of  cerebrospinal  fluid  α-­‐synuclein  in  Parkinson's  disease:  A  systematic  review  and  meta-­‐analysis.  Mov  Disord  32,  1389-­‐1400,  doi:10.1002/mds.27110  (2017).  

27   Mollenhauer,  B.  et  al.  Longitudinal  CSF  biomarkers  in  patients  with  early  Parkinson  disease  and  healthy  controls.  Neurology  89,  1959-­‐1969,  doi:10.1212/WNL.0000000000004609  (2017).  

28   Førland,  M.  G.  et  al.  Evolution  of  cerebrospinal  fluid  total  α-­‐synuclein  in  Parkinson's  disease.  Parkinsonism  Relat  Disord  49,  4-­‐8,  doi:10.1016/j.parkreldis.2018.01.018  (2018).  

29   Majbour,  N.  K.  et  al.  Longitudinal  changes  in  CSF  alpha-­‐synuclein  species  reflect  Parkinson's  disease  progression.  Mov  Disord  31,  1535-­‐1542,  doi:10.1002/mds.26754  (2016).  

30   Hall,  S.  et  al.  Longitudinal  Measurements  of  Cerebrospinal  Fluid  Biomarkers  in  Parkinson's  Disease.  Mov  Disord,  doi:10.1002/mds.26578  (2016).  

31   Stewart,  T.  et  al.  Cerebrospinal  Fluid  α-­‐Synuclein  Predicts  Cognitive  Decline  in  Parkinson  Disease  Progression  in  the  DATATOP  Cohort.  Am  J  Pathol  184,  966-­‐975,  doi:10.1016/j.ajpath.2013.12.007  (2014).  

32   Hall,  S.  et  al.  CSF  biomarkers  and  clinical  progression  of  Parkinson  disease.  Neurology  84,  57-­‐63,  doi:10.1212/WNL.0000000000001098  (2015).  

33   Paleologou,  K.  E.  et  al.  Detection  of  elevated  levels  of  soluble  alpha-­‐synuclein  oligomers  in  post-­‐mortem  brain  extracts  from  patients  with  dementia  with  Lewy  bodies.  Brain  132,  1093-­‐1101,  doi:10.1093/brain/awn349  (2009).  

34   Winner,  B.  et  al.  In  vivo  demonstration  that  alpha-­‐synuclein  oligomers  are  toxic.  Proc  Natl  Acad  Sci  U  S  A  108,  4194-­‐4199,  doi:10.1073/pnas.1100976108  (2011).  

  32  

35   Tokuda,  T.  et  al.  Detection  of  elevated  levels  of  α-­‐synuclein  oligomers  in  CSF  from  patients  with  Parkinson  disease.  Neurology  75,  1766-­‐1772,  doi:10.1212/WNL.0b013e3181fd613b  (2010).  

36   Park,  M.  J.,  Cheon,  S.  M.,  Bae,  H.  R.,  Kim,  S.  H.  &  Kim,  J.  W.  Elevated  levels  of  α-­‐synuclein  oligomer  in  the  cerebrospinal  fluid  of  drug-­‐naïve  patients  with  Parkinson's  disease.  J  Clin  Neurol  7,  215-­‐222,  doi:10.3988/jcn.2011.7.4.215  (2011).  

37   Aasly,  J.  O.  et  al.  Elevated  levels  of  cerebrospinal  fluid  α-­‐synuclein  oligomers  in  healthy  asymptomatic  LRRK2  mutation  carriers.  Front  Aging  Neurosci  6,  248,  doi:10.3389/fnagi.2014.00248  (2014).  

38   Parnetti,  L.  et  al.  Differential  role  of  CSF  alpha-­‐synuclein  species,  tau,  and  Aβ42  in  Parkinson's  Disease.  Front  Aging  Neurosci  6,  53,  doi:10.3389/fnagi.2014.00053  (2014).  

39   Hansson,  O.  et  al.  Levels  of  cerebrospinal  fluid  α-­‐synuclein  oligomers  are  increased  in  Parkinson's  disease  with  dementia  and  dementia  with  Lewy  bodies  compared  to  Alzheimer's  disease.  Alzheimers  Res  Ther  6,  25,  doi:10.1186/alzrt255  (2014).  

40   Orrú,  C.  D.  et  al.  Rapid  and  sensitive  RT-­‐QuIC  detection  of  human  Creutzfeldt-­‐Jakob  disease  using  cerebrospinal  fluid.  MBio  6,  doi:10.1128/mBio.02451-­‐14  (2015).  

41   Fairfoul,  G.  et  al.  Alpha-­‐synuclein  RT-­‐QuIC  in  the  CSF  of  patients  with  alpha-­‐synucleinopathies.  Ann  Clin  Transl  Neurol  3,  812-­‐818,  doi:10.1002/acn3.338  (2016).  

42   Groveman,  B.  R.  et  al.  Rapid  and  ultra-­‐sensitive  quantitation  of  disease-­‐associated  α-­‐synuclein  seeds  in  brain  and  cerebrospinal  fluid  by  αSyn  RT-­‐QuIC.  Acta  Neuropathol  Commun  6,  7,  doi:10.1186/s40478-­‐018-­‐0508-­‐2  (2018).  

43   Oueslati,  A.  Implication  of  Alpha-­‐Synuclein  Phosphorylation  at  S129  in  Synucleinopathies:  What  Have  We  Learned  in  the  Last  Decade?  J  Parkinsons  Dis  6,  39-­‐51,  doi:10.3233/JPD-­‐160779  (2016).  

44   Tenreiro,  S.,  Eckermann,  K.  &  Outeiro,  T.  F.  Protein  phosphorylation  in  neurodegeneration:  friend  or  foe?  Front  Mol  Neurosci  7,  42,  doi:10.3389/fnmol.2014.00042  (2014).  

45   Wang,  Y.  et  al.  Phosphorylated  α-­‐synuclein  in  Parkinson's  disease.  Sci  Transl  Med  4,  121ra120,  doi:10.1126/scitranslmed.3002566  (2012).  

46   Stewart,  T.  et  al.  Phosphorylated  α-­‐synuclein  in  Parkinson's  disease:  correlation  depends  on  disease  severity.  Acta  Neuropathol  Commun  3,  7,  doi:10.1186/s40478-­‐015-­‐0185-­‐3  (2015).  

47   Mollenhauer,  B.  et  al.  α-­‐Synuclein  and  tau  concentrations  in  cerebrospinal  fluid  of  patients  presenting  with  parkinsonism:  a  cohort  study.  Lancet  Neurol  10,  230-­‐240,  doi:10.1016/S1474-­‐4422(11)70014-­‐X  (2011).  

48   Parnetti,  L.          (Frontiers  in  Aging  Neuroscience,  2014).  49   Havelund,  J.  F.,  Heegaard,  N.  H.  H.,  Færgeman,  N.  J.  K.  &  Gramsbergen,  J.  B.  Biomarker  

Research  in  Parkinson's  Disease  Using  Metabolite  Profiling.  Metabolites  7,  doi:10.3390/metabo7030042  (2017).  

50   Yu,  Z.  et  al.  The  significance  of  uric  acid  in  the  diagnosis  and  treatment  of  Parkinson  disease:  An  updated  systemic  review.  Medicine  (Baltimore)  96,  e8502,  doi:10.1097/MD.0000000000008502  (2017).  

51   Wen,  M.  et  al.  Serum  uric  acid  levels  in  patients  with  Parkinson's  disease:  A  meta-­‐analysis.  PLoS  One  12,  e0173731,  doi:10.1371/journal.pone.0173731  (2017).  

  33  

52   Moors,  T.  et  al.  Lysosomal  Dysfunction  and  α-­‐Synuclein  Aggregation  in  Parkinson's  Disease:  Diagnostic  Links.  Mov  Disord  31,  791-­‐801,  doi:10.1002/mds.26562  (2016).  

53   Balducci,  C.  et  al.  Lysosomal  hydrolases  in  cerebrospinal  fluid  from  subjects  with  Parkinson's  disease.  Mov  Disord  22,  1481-­‐1484,  doi:10.1002/mds.21399  (2007).  

54   Parnetti,  L.  et  al.  Cerebrospinal  fluid  lysosomal  enzymes  and  alpha-­‐synuclein  in  Parkinson's  disease.  Mov  Disord  29,  1019-­‐1027,  doi:10.1002/mds.25772  (2014).  

55   Farotti,  L.,  Paciotti,  S.,  Tasegian,  A.,  Eusebi,  P.  &  Parnetti,  L.  Discovery,  validation  and  optimization  of  cerebrospinal  fluid  biomarkers  for  use  in  Parkinson's  disease.  Expert  Rev  Mol  Diagn  17,  771-­‐780,  doi:10.1080/14737159.2017.1341312  (2017).  

56   Holmberg,  B.,  Johnels,  B.,  Ingvarsson,  P.,  Eriksson,  B.  &  Rosengren,  L.  CSF-­‐neurofilament  and  levodopa  tests  combined  with  discriminant  analysis  may  contribute  to  the  differential  diagnosis  of  Parkinsonian  syndromes.  Parkinsonism  Relat  Disord  8,  23-­‐31  (2001).  

57   Abdo,  W.  F.,  Bloem,  B.  R.,  Van  Geel,  W.  J.,  Esselink,  R.  A.  &  Verbeek,  M.  M.  CSF  neurofilament  light  chain  and  tau  differentiate  multiple  system  atrophy  from  Parkinson's  disease.  Neurobiol  Aging  28,  742-­‐747,  doi:10.1016/j.neurobiolaging.2006.03.010  (2007).  

58   Constantinescu,  R.,  Rosengren,  L.,  Johnels,  B.,  Zetterberg,  H.  &  Holmberg,  B.  Consecutive  analyses  of  cerebrospinal  fluid  axonal  and  glial  markers  in  Parkinson's  disease  and  atypical  Parkinsonian  disorders.  Parkinsonism  Relat  Disord  16,  142-­‐145,  doi:10.1016/j.parkreldis.2009.07.007  (2010).  

59   Hall,  S.  et  al.  Accuracy  of  a  panel  of  5  cerebrospinal  fluid  biomarkers  in  the  differential  diagnosis  of  patients  with  dementia  and/or  parkinsonian  disorders.  Arch  Neurol  69,  1445-­‐1452,  doi:10.1001/archneurol.2012.1654  (2012).  

60   Hansson,  O.  et  al.  Blood-­‐based  NfL:  A  biomarker  for  differential  diagnosis  of  parkinsonian  disorder.  Neurology  88,  930-­‐937,  doi:10.1212/WNL.0000000000003680  (2017).  

61   Eidson,  L.  N.  et  al.  Candidate  inflammatory  biomarkers  display  unique  relationships  with  alpha-­‐synuclein  and  correlate  with  measures  of  disease  severity  in  subjects  with  Parkinson's  disease.  J  Neuroinflammation  14,  164,  doi:10.1186/s12974-­‐017-­‐0935-­‐1  (2017).  

62   Qin,  X.  Y.,  Zhang,  S.  P.,  Cao,  C.,  Loh,  Y.  P.  &  Cheng,  Y.  Aberrations  in  Peripheral  Inflammatory  Cytokine  Levels  in  Parkinson  Disease:  A  Systematic  Review  and  Meta-­‐analysis.  JAMA  Neurol  73,  1316-­‐1324,  doi:10.1001/jamaneurol.2016.2742  (2016).  

63   Lindqvist,  D.  et  al.  Cerebrospinal  fluid  inflammatory  markers  in  Parkinson's  disease-­‐-­‐associations  with  depression,  fatigue,  and  cognitive  impairment.  Brain  Behav  Immun  33,  183-­‐189,  doi:10.1016/j.bbi.2013.07.007  (2013).  

64   Brockmann,  K.  et  al.  Inflammatory  profile  discriminates  clinical  subtypes  in  LRRK2-­‐associated  Parkinson's  disease.  Eur  J  Neurol  24,  427-­‐e426,  doi:10.1111/ene.13223  (2017).  

65   Karpenko,  M.  N.,  Vasilishina,  A.  A.,  Gromova,  E.  A.,  Muruzheva,  Z.  M.  &  Bernadotte,  A.  Interleukin-­‐1β,  interleukin-­‐1  receptor  antagonist,  interleukin-­‐6,  interleukin-­‐10,  and  tumor  necrosis  factor-­‐α  levels  in  CSF  and  serum  in  relation  to  the  clinical  diversity  of  

  34  

Parkinson's  disease.  Cell  Immunol  327,  77-­‐82,  doi:10.1016/j.cellimm.2018.02.011  (2018).  

66   Schrag,  A.,  Siddiqui,  U.  F.,  Anastasiou,  Z.,  Weintraub,  D.  &  Schott,  J.  M.  Clinical  variables  and  biomarkers  in  prediction  of  cognitive  impairment  in  patients  with  newly  diagnosed  Parkinson's  disease:  a  cohort  study.  Lancet  Neurol  16,  66-­‐75,  doi:10.1016/S1474-­‐4422(16)30328-­‐3  (2017).  

67   Goldman,  J.  G.  &  Litvan,  I.  Mild  cognitive  impairment  in  Parkinson's  disease.  Minerva  Med  102,  441-­‐459  (2011).  

68   Arai,  Y.  et  al.  Alpha-­‐synuclein-­‐positive  structures  in  cases  with  sporadic  Alzheimer's  disease:  morphology  and  its  relationship  to  tau  aggregation.  Brain  Res  888,  287-­‐296  (2001).  

69   Iseki,  E.  et  al.  Dementia  with  Lewy  bodies  from  the  perspective  of  tauopathy.  Acta  Neuropathol  105,  265-­‐270,  doi:10.1007/s00401-­‐002-­‐0644-­‐3  (2003).  

70   Parnetti,  L.  et  al.  Cerebrospinal  fluid  biomarkers  in  Parkinson  disease.  Nat  Rev  Neurol  9,  131-­‐140,  doi:10.1038/nrneurol.2013.10  (2013).  

71   Compta,  Y.  et  al.  Cerebrospinal  tau,  phospho-­‐tau,  and  beta-­‐amyloid  and  neuropsychological  functions  in  Parkinson's  disease.  Mov  Disord  24,  2203-­‐2210,  doi:10.1002/mds.22594  (2009).  

72   Alves,  G.  et  al.  CSF  amyloid-­‐beta  and  tau  proteins,  and  cognitive  performance,  in  early  and  untreated  Parkinson's  disease:  the  Norwegian  ParkWest  study.  J  Neurol  Neurosurg  Psychiatry  81,  1080-­‐1086,  doi:10.1136/jnnp.2009.199950  (2010).  

73   Siderowf,  A.  et  al.  CSF  amyloid  {beta}  1-­‐42  predicts  cognitive  decline  in  Parkinson  disease.  Neurology  75,  1055-­‐1061,  doi:10.1212/WNL.0b013e3181f39a78  (2010).  

74   Terrelonge,  M.,  Marder,  K.  S.,  Weintraub,  D.  &  Alcalay,  R.  N.  CSF  β-­‐Amyloid  1-­‐42  Predicts  Progression  to  Cognitive  Impairment  in  Newly  Diagnosed  Parkinson  Disease.  J  Mol  Neurosci  58,  88-­‐92,  doi:10.1007/s12031-­‐015-­‐0647-­‐x  (2016).  

75   Vaikath,  N.  N.  et  al.  Generation  and  characterization  of  novel  conformation-­‐specific  monoclonal  antibodies  for  α-­‐synuclein  pathology.  Neurobiol  Dis  79,  81-­‐99,  doi:10.1016/j.nbd.2015.04.009  (2015).  

76   Shahnawaz,  M.  et  al.  Development  of  a  Biochemical  Diagnosis  of  Parkinson  Disease  by  Detection  of  α-­‐Synuclein  Misfolded  Aggregates  in  Cerebrospinal  Fluid.  JAMA  Neurol  74,  163-­‐172,  doi:10.1001/jamaneurol.2016.4547  (2017).  

77   del  Campo,  M.  et  al.  Recommendations  to  standardize  preanalytical  confounding  factors  in  Alzheimer's  and  Parkinson's  disease  cerebrospinal  fluid  biomarkers:  an  update.  Biomark  Med  6,  419-­‐430,  doi:10.2217/bmm.12.46  (2012).  

78   Mollenhauer,  B.  et  al.  A  user's  guide  for  α-­‐synuclein  biomarker  studies  in  biological  fluids:  Perianalytical  considerations.  Mov  Disord  32,  1117-­‐1130,  doi:10.1002/mds.27090  (2017).  

   

  35    

!!

CHAPTER 2

Generation and Characterization of Novel Conformation-Specific Monoclonal Antibodies for α-Synuclein Pathology

  36  

CHAPTER 2 | GENERATION AND CHARACTERIZATION OF NOVEL CONFORMATION-SPECIFIC MONOCLONAL ANTIBODIES FOR Α-SYNUCLEIN PATHOLOGY Nishant N. Vaikath1, 2, Nour K. Majbour1,3, Katerina E. Paleologou4, Mustafa T. Ardah1, Esther van Dam4, Wilma D. J. van de Berg3, Shelley L. Forrest5, Laura Parkkinen6, Wei-Ping Gai7, Nobutaka Hattori8, 9, Masashi Takanashi9, Seung-Jae Lee10, David M. A. Mann11, Yuzuru Imai8, Glenda M Halliday12, Jia-Yi. Li2, Omar M. A. El-Agnaf1, 13

1Department of Biochemistry, College of Medicine and Health Science, United Arab Emirates University, Al Ain, United Arab Emirates 2Neural Plasticity and Repair Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, BMC A10, Lund University, Lund, Sweden. 3Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands. 4Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece 5Discipline of Pathology, Charles Perkin Centre, University of Sydney, Sydney, Australia 6Department of Clinical Neurology, University of Oxford, Oxford, UK 7Department of Human Physiology, School of Medicine, Flinders University, Australia 8Department of Research for Parkinson’s Disease, Juntendo University Graduate School of Medicine, Japan 9Department of Neurology, Juntendo University Graduate School of Medicine, Japan 10Department of Biomedical Science and Technology, Konkuk Univeristy, Seoul 143-701, Korea 11Clinical and Cognitive Neuroscience Research Group, University of Manchester, Salford Royal Foundation NHS Trust, Salford M6 8HD, UK 12Faculty of Medicine, University of New South Wales and Neuroscience Research Australia, Sydney, Australia 13College of Science, Engineering and Technology, HBKU, Education City, Qatar Foundation, Doha, Qatar Neurobiol Dis. 2015 Jul;79:81-99. doi: 10.1016/j.nbd.2015.04.009..

 

 

 

 

 

 

 

  37  

Abstract α-Synuclein (α-syn), a small protein that has the intrinsic propensity to aggregate, is implicated in several

neurodegenerative diseases including Parkinson’s disease (PD), dementia with Lewy bodies (DLB), and

multiple system atrophy (MSA), which are collectively known as synucleinopathies. Genetic, pathological,

biochemical, and animal modeling studies provided compelling evidence that α-syn aggregation plays a

key role in the pathogenesis of PD and related synucleinopathies. It is therefore of utmost importance to

develop reliable tools that can detect the aggregated forms of α-syn. We describe here the generation and

characterization of six novel conformation-specific monoclonal antibodies that recognize specifically α-

syn aggregates but not the soluble, monomeric form of the protein. The antibodies described herein did not

recognize monomers or fibrils generated from other amyloidogenic proteins including β-syn, γ-syn, β-

amyloid, tau protein, islet amyloid polypeptide and ABri. Interestingly, the antibodies did not react to

overlapping linear peptides spanning the entire sequence of α-syn, confirming further that they only detect

α-syn aggregates. In immunohistochemical studies, the new conformation-specific monoclonal antibodies

showed underappreciated small micro-aggregates and very thin neurites in PD and DLB cases that were not

observed with generic pan antibodies that recognize linear epitope. Furthermore, employing one of our

conformation-specific antibodies in a sandwich based ELISA, we observed an increase in levels of α-syn

oligomers in brain lysates from DLB compared to Alzheimer’s disease and control samples. Therefore, the

conformation-specific antibodies portrayed herein represent useful tools for research, biomarkers

development, diagnosis and even immunotherapy for PD and related pathologies.

Introduction

The fibrillization of α-synuclein (α-syn) is considered to play a key role in the pathogenesis of several

neurodegenerative diseases including Parkinson’s disease (PD), dementia with Lewy bodies (DLB), and

multiple system atrophy (MSA) [1]. Indeed, many histopathological studies have demonstrated that α-syn

fibrils are the main components of the neuronal inclusions, namely Lewy bodies (LBs) and Lewy neurites

(LNs), which are the pathological hallmarks of PD and DLB, and glial cytoplasmic inclusions (GCIs), the

pathological hallmark of MSA [2-4]. Genetic studies show various point mutations (A53T, A30P, E46K,

H50Q or G51D) [5-9] and multiplication of the α-syn gene SNCA lead to familial PD [10, 11]. Cell culture

and animal model studies indicate that enhanced oligomerization and aggregation of α-syn is associated

with increased cytotoxicity [12-16], while α-syn oligomers have been shown to be significantly elevated in

the brain lysates and cerebrospinal fluid of PD and DLB patients [17-23]. The significance of α-syn

fibrillization in the pathobiology of synucleinopathies has been reinforced by recent data demonstrating

that preformed recombinant α-syn fibrils can seed the aggregation of endogenous α-syn both in cultured

  38  

wild-type mouse hippocampal neurons [24] and in in vivo transgenic and wild-type mice [25, 26], even

from the peripheral to the central nervous system [27].

Given the central role of α-syn in the pathogenesis of synucleinopathies, the characterization of

their pathology in the brain has relied heavily on the use of α-syn antibodies [4, 28, 29], the vast majority

of which recognize both the monomeric and the aggregated forms of α-syn. Hence, there is a need for

developing conformation-specific antibodies that can possibly unveil underappreciated α-syn

neuropathology or even reveal novel neuropathological features of PD and related disorders. Furthermore,

taking into account that passive immunotherapy against α-syn has emerged as a very promising strategy for

modifying PD and related synucleinopathies [30-32], therefore, the generation of conformation-specific

antibodies may prove to be a very useful tool for the treatment of these diseases.

The aim of the present study was to generate and characterize monoclonal antibodies (mAbs) that

specifically target the aggregated forms of α-syn. The specificity and sensitivity of these antibodies were

assessed by an array of biochemical methods including dot-blotting, ELISA, BIACORE and

immunohistochemical analysis.

Materials and methods Expression and purification of recombinant human α-syn

Expression and purification of recombinant human α-syn was performed as previously described [33].

Briefly, the glutathione S-transferase (GST)-α-syn fusion construct in pGEX-4T1 vector was inserted into

E.coli BL21 bacteria for protein expression. The transformed bacteria were grown in Luria Bertani (LB)

medium, and glutathione sepharose 4B beads were used to capture GST-α-syn in the bacterial lysate, and

the GST tag was cleaved by thrombin. The molecular weight and purity of α-syn was confirmed by SDS-

PAGE gel and the concentration was estimated using a BCA protein assay (Pierce Biotechnology,

Rockford, IL). For the expression of recombinant truncated human α-syn (1-122), the construct (kindly

provided by Dr. Hilal A. Lashuel, EPFL, Lausanne, Switzerland) was inserted into a bacterial expression

vector (pGEX-4T1 vector) and purification carried out as mentioned above.

Preparation of fibrils of α-syn and other amyloid proteins and peptides

Recombinant α-, β- or γ-syn (50µM) in phosphate buffered saline (PBS) were incubated in a thermomixer

(800 rpm) at 37°C. The incubation period was 7 days for α-syn and 15 days for β- and γ-syn. To prepare

pure fibrils, aggregated α-syn solution for seven days was spun at 10,000 x g for 10 minutes at 4°C in a

refrigerated microfuge (Eppendorf). The supernatant was then discarded, and the pellet was washed twice

with ultra-pure water, before resuspended in 1x PBS. The pure fibrils were fragmented by ultrasonication

while kept on ice using a Sonic ruptor 250, equipped with a fine tip (5 sec pulses, output of 40 watts for 5

minutes). For the estimation of α-syn concentration of the fibrils, the samples were denatured by 6M

Guanidine-HCl and BCA assay (pierce BCA protein assay kit; Thermo scientific) were used according to

  39  

manufacturer’s instructions. Aggregates of other amyloid proteins and peptides were prepared as previously

described. The non-amyloid component of α-syn (NAC) (61-95), NAC (61-78) and β-amyloid (Aβ)

peptides were prepared as previously reported [34]. Tau40 (kindly provided by Prof. Dominic Walsh,

Harvard Medical School) was aggregated as described previously [35]. Islet amyloid polypeptide (IAPP)

and ABri were prepared as previously reported [36, 37]. The fibrillization of α-syn and other amyloid

protein and peptides was monitored by Thioflavin-S (Th-S) binding assay and transmission electron

microscopy (TEM).

Thioflavin-S (Th-S) assay

10 µl of each sample was diluted in 40 µl of Th-S (25 µM) in PBS. Fluorescence was then measured using

a Victor X32030 multilabel plate reader (Perkin Elmer, USA) with the excitation and emission wavelengths

set at 450 and 486 nm, respectively. To avoid thebackground fluorescence, the fluorescence intensity of a

blank PBS containing Th-S was subtracted from all readings.

Transmission electron microscopy (TEM)

The samples (5 µl) were deposited on Formvar-coated 400-mesh copper grids (Agar Scientific, UK), fixed

briefly with 0.5% glutaraldehyde (5 µl), negatively stained with 2% uranyl acetate (Sigma–Aldrich, USA)

and examined with a Philips CM-10 TEM electron microscope.

Generation and purification of anti-α-syn monoclonal antibodies

The experimental procedures using mice were carried out in accordance with the UAE University ethical

procedure for experimental animals and approved by the Animal Research Ethics Committee, UAE

University (No. A24-13). 6-8 weeks old Balb/c female mice were immunized by subcutaneous injection

with α-syn fibrils (50 µg/mouse) emulsified in complete Freund’s adjuvant (Sigma-Aldrich, USA) followed

by booster immunization (25 µg/mouse) in incomplete Freund’s adjuvant (Sigma-Aldrich, USA) after a

three-week interval. Mice with high antibody titers received a final intraperitoneal injection of α-syn fibrils

(150 µg/mouse) in PBS. Four days after the final injection, mice were sacrificed and their spleen cells were

fused with Sp20 myeloma cells using PEG 4000 (Merck, Germany). The fused cells were selected for

hybridomas using Iscove’s Modified Dulbecco’s Medium (IMDM) (Gibco, Rockville, MD, USA)

supplemented with hypoxanthine-aminopterin-thymidine (HAT) (Sigma-Aldrich, USA). Indirect ELISA

was carried out to screen the antibody-producing hybridomas, which were sub-cloned and passaged at least

three times to ensure monoclonality, before they were expanded in serum free media (CDM4mAb, Hyclone

Laboratories, Logan, UT, USA) for antibody purification. The isotypes of the antibodies were determined

using an isotyping kit (Sigma-Aldrich, USA). The antibodies were purified by Protein G-agarose

(Calbiochem) affinity chromatography.

  40  

Indirect ELISA

A 96-well clear maxisorp plate (Nunc, Denmark) was coated with 70 ng/well of α-syn fibrils in PBS and

was incubated at 4°C overnight. The plate was then washed three times with PBS-T(PBS containing 0.05%

Tween 20) and incubated with blocking buffer (PBS-T containing 2.25% gelatin) for 1 h at RT. After an

additional wash with PBS-T, 100 µl of antiserum (for titer check) or hybridoma culture supernatant (for

screening of positive clones) was dispensed in each well and the plate was incubated for 1 h at RT. The

plate was then washed with PBS-T and incubated with HRP-conjugated goat anti-mouse secondary

antibody (Jackson ImmunoResearch Laboratories Inc., USA) for 1 h at RT. After washing with PBS-T,

50µl/well of tetramethylbenzidine (TMB) peroxidase substrate (KPL, Maryland, USA) was added. The

reaction was stopped by adding 0.6 N H2SO4 (100 µl/well) and the absorbance at 450nm was measured in

a Victor X3 2030 multilabel plate reader (Perkin Elmer, USA).

Inhibition ELISA

A 384-well black maxisorp plate (Nunc, Denmark) was coated with 1.4 µg/ml of α-syn fibrils (50 µl/well)

in PBS and incubated at 4°C overnight. Blocking was carried out as described for the indirect ELISA. α-

Syn fibrils or monomers at starting concentration of 80 µM were subjected to two-fold serial dilution in

blocking buffer generating 18 different concentrations in 0.6 ml siliconized tubes. Equal volume of 20

ng/ml of our mAbs (Syn-F1, Syn-F2, Syn-O1, Syn-O2, Syn-O3 and Syn-O4) or the control antibody (Syn-

1) were added to each dilution and mixed for 2.5 h at RT. The antibody-antigen mixture was then added to

the antigen-coated–plate and incubated for 10 minutes at RT. Following washes with PBS-T, HRP-

conjugated goat anti-mouse secondary antibody (1:15000; Jackson ImmunoResearch Laboratories Inc.,

USA) was added and incubated for 1h at RT. The bound HRP activities were assayed by chemiluminescent

reaction using 50 µl/well of an enhanced chemiluminescent substrate (SuperSignal ELISA Femto

Maximum Sensitivity Substrate, Pierce Biotechnology, Rockford, USA). The chemiluminescence in

relative light units was then measured in a Victor X32030 multi-label plate reader (Perkin Elmer, USA).

Dot Blot

Nitrocellulose membrane was spotted with 50 ng (5 µl/spot) of protein and blocked with blocking buffer

(5% skimmed milk in PBS-T) for 1 h at RT. After washing, the blots were probed with the generated

antibodies or control antibodies (see Table. 3) specific for Aβ (82E1, IBL America, 50ng/ml), tau (5E2,

kindly provided by prof. Dominic Walsh, Harvard Medical School, 50ng/ml,), IAPP (R10/99, Santa Cruz

Biotechnology, 100ng/ml), ABri (Rabbit anti-ABri antiserum, kindly provided by Prof. Dominic Walsh,

Harvard Medical School, 1/3000), β-syn (Anti-beta synuclein (8), Santa Cruz Biotechnology, 1:2000), γ-

syn (C-20, Santa Cruz Biotechnology, 1:3000) α-syn (Syn-1, BD Bioscience, 50ng/ml), truncated α-syn (1-

122), NAC(61-95) and NAC(61-78) (5C2, Santa Cruz Biotechnology, 1:50), and incubated for 2 h at RT.

For the pre-absorption experiment, the antibodies were mixed with 5µg/ml of either α-syn fibrils or

  41  

monomers for 2 h at RT and then incubated with the nitrocellulose membrane, which was spotted with α-

syn fibrils or monomers. The blots were washed and incubated with the appropriate HRP-conjugated

secondary antibody for 1 h at RT and developed with chemiluminescent substrate (SuperSignal west pico

substrate, Pierce Biotechnology, Rockford, USA).

Determination of mAbs affinities by surface plasmon resonance

The kinetic constants for the interaction between the six raised mAbs and α-syn fibrils were determined by

surface plasmon resonance on a BIAcore X 100 instrument (Biacore, GE Healthcare). CM5 sensor chip

was activated with N-hydroxysuccinimide (NHS) and 1-ethyl-1-(dimethylaminopropyl)-carbofiimide

(EDC). α-Syn fibrils were immobilized on the chip by injecting 35 µl (120 µg/ml) of α-syn fibrils in 10

mM sodium acetate buffer, pH 4.5. The association rate constants were obtained by injecting the raised

antibodies or the control antibody, anti-α-syn 211 (Santa Cruz Biotechnolgy, USA) at eight different

concentrations ranging from 0.01 to 10 nM in HBS running buffer (10 mM HEPES, 150 mM NaCl, 3.4

mM EDTA and 0.005% surfactant P20, pH 7.4) at a flow rate of 10 µl/minute. The dissociation rates were

measured at a flow rate of 40 µl/min. The sensor chip was regenerated using 100 mM NaHCO3, pH. 9.6

and the sensograms analyzed with the BOA evaluation software (Biacore, GE Healthcare).

Epitope mapping

Synthetic fourteen amino acid long peptides with 7 amino acid overlap, covering the entire sequence of α-

syn (see Table 2) were used (Shanghai Hanhong Chemical Co., China). A 384-well black maxisorp plate

(Nunc, Denmark) was coated with 500 ng/well of the peptides in 0.2 M NaHCO3, pH 9.6 or 70 ng/well of

α-syn fibrils in PBS, pH 7.4 and incubated uncovered at 37°C overnight to dry out. The plate was washed

with PBS-T and blocked with 100 µl/well of blocking buffer (2.25% gelatin in PBST) for 1 h at RT.

Subsequent to washes with PBS-T, 50 µl/well (100 ng/ml) of our antibodies was added and the plate was

incubated for 1h at RT. Syn-1 (200 ng/ml, mouse anti-α-syn; BD Bioscience), N-19 (200 ng/ml, goat anti-

α-syn; Santa Cruz Biotechnology), FL-140 (200 ng/ml, rabbit anti-α-syn; Santa Cruz Biotechnology), 5C2

(1 µg/ml, mouse anti-α-syn; Santa Cruz Biotechnology) and 3B6 (1 µg/ml, mouse anti-α/β-syn; Santa Cruz

Biotechnology) were also included as control antibodies (see Table 3). After incubation with the appropriate

secondary antibody, HRP conjugated goat anti-mouse IgG (1:20000; Jackson ImmunoResearch

Laboratories Inc., USA), goat anti-rabbit IgG (1:5000; Jackson ImmunoResearch Laboratories Inc., USA),

or chicken anti-goat IgG (1:20000; Jackson ImmunoResearch Laboratories Inc., USA) for 1 h at RT, the

plate was incubated with 50 µl/well of substrate (Super Signal ELISA Femto Maximum Sensitivity

Substrate, Pierce Biotechnology, Rockford, USA) and the chemiluminescence in relative light units

measured with a Victor X3 2030 multi-label plate reader (Perkin Elmer, USA). The ELISA plates were

then exposed onto x-ray film and then developed to obtain the pepscan blots.

  42  

Table 1. Comparison of binding affinity of conformation-specific mAbs !

mAbs Isotype

Binding affinity by Inhibition ELISA

Kinetic constants by Surface Plasmon Resonance

IC50 (mean ±SD) Fold

affinity to

fibrils

ka (1/Ms)

kd (1/s)

KD (M) Fibrils

(µM)

Monomers (µM)

Syn-F1

IgG1

0.00012 ±2.1x10-6

2.083 ±0.103

16558 1.23 x 107 1.56 x 10-3 1.27 x 10-10

Syn-F2 IgG2a 0.00118

±1.4x10-5

3.707 ±0.237 3141 1.25 x 106 3.37 x 10-3 2.68 x 10-9

Syn-O1 IgG2b 0.00037

±5.3x10-5

4.793 ±0.785 12737 3.59 x 106 6.93 x 10-4 1.92 x 10-10

Syn-O2 IgG1 0.00012

±4.4x10-6

3.283 ±0.212 27312 1.31 x 107 1.27 x 10-3 9.6 x 10-11

Syn-O3 IgG1 0.00047

±6.3x10-4

5.329 ±1.75 11129 1.78 x 106 4.8 x 10-4 2.76 x 10-10

Syn-O4 IgG1 0.00052

±7.7x10-6

3.605 ±0.111 6932 7.23 x 106 9.64 x 10-4 1.37 x 10-10

Syn-1 IgG1 0.00210 ±2.1x10-5

0.001 ±3.5x10-5 0.476 - - -

211 IgG1 - - - 1.64 x 105 4.22 x 10-4 2.56 x 10-9

  43  

Table 2. Sequence of α-syn peptide library. Each peptide is 14 amino acid long (7 amino acid overlap) spanning the sequence of human α-syn.

Peptide No.

Sequence No.

Peptide sequence

1 1-14 MDVFMKGLSKAKEG 2 8-21 LSKAKEGVVAAAEK 3 15-28 VVAAAEKTKQGVAE 4 22-35 TKQGVAEAAGKTKE 5 29-42 AAGKTKEGVLYVGS 6 36-49 GVLYVGSKTKEGVV 7 43-56 KTKEGVVHGVATVA 8 50-63 HGVATVAEKTKEQV 9 57-70 EKTKEQVTNVGGAV 10 64-77 TNVGGAVVTGVTAV 11 71-84 VTGVTAVAQKTVEG 12 78-91 AQKTVEGAGSIAAA 13 85-98 AGSIAAATGFVKKD 14 92-105 TGFVKKDQLGKNEE 15 99-112 QLGKNEEGAPQEGI 16 106-119 GAPQEGILEDMPVD 17 113-126 LEDMPVDPDNEAYE 18 120-133 PDNEAYEMPSEEGY 19 127-140 MPSEEGYQDYEPEA

Time dependent aggregation of α-syn

Fresh α-syn solution (50 µM) in PBS was incubated at 37°C with continuous mixing (800 rpm) in a

thermomixer. Aliquots of the incubated solution were collected at different time points (0, 2, 4, 6, 16, 24,

36 and 48 h), flash frozen with liquid nitrogen and stored at -80°C until required for dot blotting or sandwich

ELISA. Aggregation was monitored by Th-S assay, western blotting and transmission TEM.

Western blotting

The samples (30 ng) collected at various time points during the α-syn aggregation process were mixed with

the non-reducing sample buffer (0.25 M Tris-HCl, pH 6.8, 50% glycerol, 0.01% bromophenol blue), loaded

into a 15 % SDS-PAGE gel and separated by electrophoresis at 100V for 90 minutes. The use of non-

denaturing condition helps to maintain the high molecular weight species present in aggregated samples

compared with the samples prepared in denaturing condition [38]. The proteins were subsequently

  44  

Table 3. List of commercial antibodies used in this study.

Sl. No.

Antibody

Source

Epitope

Experiment

Dilution

1 Amyloid β, (82E1)

IBL America - Dot blot 50 ng/ml

2 Anti Tau antibody, 5E2

Gift from Prof. Dominic Walsh, Harvard medical

school

- Dot blot 50 ng/ml

3 Amylin antibody, R10/99

Santa Cruz Biotechnology

- Dot blot 100 ng/ml

4 Anti-ABri antiserum

Gift from Prof. Dominic Walsh, Harvard medical

school

- Dot blot 1/3000

5 β-synuclein antibody (8)

Santa Cruz Biotechnology

- Dot blot 1/2000

6 γ-synuclein Antibody (C-

20)

Santa Cruz Biotechnology

- Dot blot 1/3000

7 α-synuclein antibody (Syn-1)

BD Biosciences 91-99 of α-syn

Dot blot and ELISA

200 ng/ml

8 α-synuclein antibody, 5C2

Santa Cruz Biotechnology

61-95 of α-syn

Dot blot and ELISA

1/50 (Dot Blot) and 1µg/ml

(ELISA)

9 α-synuclein antibody, N19

Santa Cruz Biotechnology

1-60 of α-syn

ELISA 200 ng/ml

10 α-synuclein antibody, FL-140

Santa Cruz Biotechnology

Full length of α-syn

ELISA and western blot

200ng/ml (ELISA)

and 1/3000

(Western blot)

11 α/β-synuclein

antibody, 3B6 Santa Cruz

Biotechnology 119-140 of α-syn

ELISA 1µg/ml (ELISA)

  45  

transferred into a nitrocellulose membrane and western transfer performed at 90V for 110 minutes. The

membrane was boiled in PBS for 5 minutes and blocked with blocking buffer (5% skimmed milk in PBS-

T) for 1 h at RT followed by incubation with α/β/γ-syn FL-140 polyclonal antibody (1:3000 in PBS-T,

Santa Cruz Biotechnology, USA) for 2 h at RT. After washing with PBS-T, the membrane was incubated

for 1 h with HRP-conjugated goat anti-rabbit IgG (1:120.000 in PBS-T, Jackson ImmunoResearch

Laboratories Inc., USA). Following an additional washing step with PBS-T, the membrane was developed

with Super signal west femtochemiluminescent substrate (Pierce Biotechnology, Rockford, USA).

ELISA for measuring α-syn aggregates

A 384-well ELISA maxisorp plate (Nunc, Denmark) was coated by overnight incubation with either 0.1

µg/ml of our mAbs or Syn-1 (BD Bioscience, San Diego) (50 µl/well) in coating buffer (200 mM NaHCO3

buffer, pH 9.6). After washing with PBS-T, the plates were incubated with blocking buffer (PBS-T

containing 2.25% gelatin and 1% BSA) for 2 h at 37°C. Aggregated α-syn collected at various time points

(0, 2, 4, 6, 16, 24, 36 and 48 h) at a concentration of 5 nM (50 µl/well in PBS) was added and incubated for

5 minutes at RT, followed by incubation with 50 µl/well of the FL-140 rabbit polyclonal antibody (Santa

Cruz Biotechnology, USA, 1:1000) for 2 h at 37 °C. Subsequently, 50 µl/well HRP-conjugated goat anti-

rabbit IgG (Jackson ImmunoResearch Laboratories Inc., USA, 1:15000) was added and incubated for 1.5

h at 37°C. The bound HRP was detected by adding 50 µl/well of substrate (Super Signal ELISA Femto

Maximum Sensitivity Substrate, Pierce Biotechnolgy, Rockford, USA) and the chemiluminescence in

relative light units was measured in a Victor X3 2030 multi-label plate reader (Perkin Elmer, USA).

ELISA for measuring of α-syn oligomers in human brain homogenates

Frozen post-mortem samples of the frontal cerebral cortex from clinically diagnosed and

neuropathologically-confirmed DLB patients (n=14), AD patients (n=9) and age-matched controls (n=7)

were obtained from Manchester Brain Bank and processed as previously reported [18]. Briefly, tissue

samples were homogenized in CelLytic buffer (Sigma-Aldrich, USA) containing a cocktail of protease

inhibitors, including AEBSF, aprotinin, E-64, EDTA and leupeptin (Calbiochem, Germany) and

centrifuged at 100,000 x g for 3 h at 4°C.. The supernatant was collected, total protein concentration was

measured and all samples were adjusted to 0.1 mg/ml and stored at -80°C.

A 384-well ELISA plate (Nunc, Denmark) was coated with 0.2 µg/ml of Syn-O2 antibody (50 µl/well), in

200 mM NaHCO3 buffer, pH 9.6, and incubated at 4°C overnight. After incubation with 100µl/well of

blocking buffer (PBS-T containing 2.25% gelatin) for 2 h at 37°C, 50 µl/well of the brain lysates (5 µg/ml

in CelLytic buffer, Sigma-Aldrich, USA) and serial dilutions of α-syn oligomers (as standard generated

from aggregation of α-syn with Gn Rb1 [38] was dispensed in each well and the plate was incubated at

37°C for 2.5 h. After washing with PBS-T, 50 µl/well of FL-140 antibody (1:1000) was added and the plate

was incubated for another 1 h at 37°C. Subsequently, 50 µl/well of HRP-conjugated anti-rabbit antibody

  46  

(Jackson ImmunoResearch Laboratories Inc., USA, 1:15000) was added and incubated for 1 h at 37°C. The

plate was then washed and incubated with 50 µl/well of an enhanced chemiluminescent substrate (Super

Signal ELISA Femto Maximum Sensitivity Substrate, Pierce Biotechnology, Rockford, USA) and the

chemiluminescence in relative light units was measured with a Victor X32030 multi-label plate reader

(Perkin Elmer, USA). The samples were screened in a blinded fashion and tested randomly in duplicates.

All results were confirmed with at least two independent experiments. A series of internal controls was also

run to check for run-to-run variations. The intra-assay and inter-assay coefficient of variation examined

over >10 plates was < 10%. The lower limit of detection was as low as 10 pg/ml. The lower limit of

quantification for the assay was determined to be 10 pg/ml and the upper limit of quantitation was 10 ng/ml.

The concentration of α-syn oligomers in the samples was calculated using the standard curve.

Immunohistochemistry with conformation-specific α-syn antibodies of human brain tissue samples

The conformation-specific anti-α-syn mAbs were tested by five independent laboratories using

immunohistochemistry: Laboratory 1 (Van de Berg, Amsterdam) evaluated the immunostaining in the

substantia nigra (SN), hippocampus and entorhinal cortex of idiopathic PD, DLB and age-matched controls;

Laboratory 2 (Parkkinen, Oxford) evaluated immunostaining using different antigen retrieval methods in a

DLB patient. Laboratory 3 (Halliday, Sydney) evaluated the immunostaining in the anterior cingulate cortex

of PD, AD and aged, neurologically normal control cases, and the putamen from a MSA case; Laboratory

4 (Imai, Tokyo) evaluated the double immunofluorescence staining for brain sections from PD and control

cases; Laboratory 5 (Gai, Flinders) employed double immunofluorescence staining to compare the staining

patternson isolated cortical LBs from DLB cases. In agreement with local ethical guidelines, informed

consent was obtained for the use of all donor brain tissue, and neuropathological and clinical information

for research purposes.

Laboratory 1 (Van de Berg, Amsterdam): Transverse 10-µm-thick sections of formalin-fixed, paraffin-

embedded brain tissue including mesencephalon, hippocampus and entorhinal cortex of clinically

diagnosed and pathologically confirmed PD, DLB and aged-matched non-demented control cases (n=4 per

group), collected by the Netherlands Brain Bank (NBB, Amsterdam, The Netherlands), were processed for

immunohistochemistry. Antigen retrieval consisted of steaming in citrate buffer pH 6.0 followed by

blocking in 5% bovine albumin (BSA) in TBS-triton and then the elimination of endogenous peroxidase

activity with treatment with 3% H2O2 in PBS. Adjacent sections were then incubated with our mAbs (Syn-

O1, Syn-O2, Syn-O3, Syn-O4, F1, F2; 200 ng/ml), Syn-1 (BD Laboratories; 125 ng/ml) and KM51

(Novocastra, Newcastle, UK; dilution 1:500, full length) overnight at 4oC. To visualize the primary

antibodies, the EnVisiontm system (DAKO) was used which contains the specific secondary antibodies for

anti-mouse lgG. Next, 3, 3’-diaminobenzidine (DAB) peroxidise (Sigma-Aldrich) was applied for antigen

visualisation and sections were counterstained using 0.5% cresyl violet.

  47  

Laboratory 2 (Parkkinen, Oxford): All six antibodies were tested with no pretreatment or with three

different antigen retrieval methods (a. autoclave at 120°C for 10 min in citrate buffer; b. 98%formic acid

for 15 min; c. 20 µg/ml of proteinase K for 5 min) in formalin-fixed, paraffin-embedded 6-µm-thick sections

from the entorhinal cortex of a pathologically confirmed DLB patient (81 years, male) collected by the

Oxford Brain Bank. Following antigen retrieval, endogenous peroxidase activity was eliminated with

treatment with 3% H2O2 (in PBS), antibodies were each diluted (200 ng/ml) and adjacent sections incubated

overnight at 4°C. For detection, Histostain SP kit (Zymed, San Francisco, CA) was used with Romulin AEC

chromogen (Biocare Medical, Walnut Creek, CA).

Laboratory 3 (Halliday, Sydney):Immunostaining was performed on 10-µm-thick, formalin-fixed,

paraffin-embedded brain sections of the anterior cingulate cortex of clinically diagnosed and pathologically

confirmed PD (mean disease duration of 13±11 y), AD (disease duration of 6 y) and aged-matched control

cases (males n=2 per group, mean group age at death 81±6 y), and from the putamen of a male MSA case

(duration of 8 years, aged 67 at death) collected by the Sydney Brain Bank. Antigen retrieval with formic

acid (90%) for 3 min was followed by blocking endogenous peroxidase activity using 1% H2O2 (in 50%

ethanol) and non-specific binding using 10% normal horse serum. Adjacent sections were incubated

overnight at 4°C with Syn-1 (BD Laboratories; 125 ng/ml), anti-phospho-S129 α-syn antibody (Abcam;

1:500), Syn-F1 (100 ng/ml), Syn-F2 (200 ng/ml), Syn-O1 (100 ng/ml), Syn-O2 (200 ng/ml), Syn-O3 (200

ng/ml) or Syn-O4 (200 ng/ml), washed and incubated with biotinylated horse anti-mouse (Vector

Laboratories, USA; 1:200) or goat anti-rabbit (Vector Laboratories, USA; 1:200) antisera for 30 minutes at

37°C, followed by avidin-biotin complex (Vectastain Elite kit; Vector Laboratories, USA; 1:500) for 30

minutes at RT. Visualization with DAB in 0.005% H2O2 was used and sections counter-stained with 0.5%

cresyl violet.

Laboratory 4 (Imai, Tokyo): Double immunofluorescence was performed using 10-µm-thick, formalin-

fixed, paraffin-embedded sections from the amygdala of clinically diagnosed and pathologically confirmed

PD and control cases collected by the Department of Neurology, Jutendo University Hospital, Japan and

approved by the ethical committee of Jutendo University. Deparaffinized sections microwaved in citrate

buffer pH 6.0 for 7 min for antigen retrieval, were neutralized with PBS pH 7.4. Sections were blocked

with 5% goat serum and incubated with primary antibodies (Syn-F2 and Syn-O4; 500 ng/ml; MJFR1; 2

µg/ml) overnight at 4˚C. The primary antibodies were visualized with Alexa Fluor488 goat anti-mouse IgG

(1:400, Life Technologies) and Alexa Fluor594 goat anti-rabbit IgG (1:400) for 2 h at RT. To reduce the

lipofuscin autofluorescence, sections were further treated with 0.1% Sudan black B (CI 26150, Wako) in

70% ethanol for 1 min and washed with PBS briefly, prior to mounting with Vectashield (H-1000, Vector

Laboratories) and assessed using a TCS-SP5 laser-scanning microscope system (Leica Microsystems). For

  48  

tissue integrity, the same brain samples were stained with DAB and Hematoxylin, and indicated the cell

morphology is well conserved (data not shown).

Laboratory 5 (Gai, Flinders): Immunofluorescence double staining was used to compare the staining

patterns of our conformation-specific mAbs with generic α-syn antibodies, on isolated cortical LBs. The

LBs were isolated from temporal cortices of two DLB cases, and smears made as described [39]. Briefly,

fresh frozen brain tissues were taken from two cases [sex/age at death/ postmortem interval (h): m/80/6,

f/81/12]. Gray matter was dissected from the temporal cortex comprising the superior, middle, inferior, and

fusiform gyri from a 1-cm-thick coronal slice at the level of caudal amygdaloid, and homogenized in four

volumes of homogenization buffer (HB; 0.32 M sucrose, 50 mM Tris-HCl at pH 7.4, 5 mM EDTA,

leupeptin 1 mg/ml, pepstatin 1 mg/ml, phenylmethylsulfonyl fluoride 17.4 mg/ml), and homogenized in a

Dounce homogenizer (Wheaton) by 10 strokes of pestle A and 10 strokes of pestle B. The homogenate was

filtered through glass wool and centrifuged for 10 min at 1000 x g. The pellet was adjusted to 6 ml with HB

and 14% (v/v) of Percoll concentration. The suspension was overlaid on 2.4 ml of 35% Percoll (v/v in HB)

and centrifuged for 30 min at 35,000 x g. The material between the sample-35%Percoll interface was

collected and washed once in TBS (50 mM Tris–HCl buffered saline pH 7.4 and protease inhibitors) by

centrifugation for 10 min at 4000 x g. The above procedures were conducted either on ice or at 4°C. The

pellet was resuspended in 10 volumes of TBS and smeared onto gelatin-coated slides and air-dried at room

temperature. The smears were fixed for 10 min with 2% paraformaldehyde with 0.2% picric acid in PBS,

followed by 3 X 5 min washes with TBS-azide (20 mM Tris-HCl, 0.15M NaCl, 0.1% sodium azide, pH

7.4), blocked for 60 min with 20% normal horse serum (NHS) in TBS-azide, incubated overnight with

primary antibody pairs constituted in 1% NHS TBS-azide. Primary antibodies were affinity-purified sheep

anti-α-syn [40] and our mAbs (1 µg/ml). Following 3 X 5 min washes with TBS-azide, the smears were

incubated for 1 h with Alexa 594 or Alexa 488 conjugated donkey anti-sheep and donkey anti-mouse

(Invitrogen) IgG. The smears were washed 3 X 5 min washes with TBS-azide, and coverslipped with 20µl

of Vectashield mounting medium, sealed and examined using a Leica TCS SP5 Spectral Confocal

Microscope.

Statistical analysis

Statistical significance for the group comparisons for the levels α-syn oligomers in human brain

homogenates was analyzed by one-way ANOVA with Dunnett post-hoc test. The data were represented as

mean. Analyses were performed using GraphPad Prism software version 5.00 (GraphPad Software, San

Deigo, CA, USA). P-value <0.05 were considered as significant.

Results Generation of mAbs specific for α-syn pathology

  49  

In order to generate mAbs specific for α-syn pathology, we immunized Balb/c mice with α-syn fibrils. We

observed that all mice showed a high antibody titer immediately after the first booster immunization and,

as a consequence, they did not receive any further immunization prior to sacrificing. By employing an

indirect ELISA, we identified 1,100 positive clones for α-syn aggregates that were subcultured at least three

times to isolate the stable ones. Using dot blotting, we identified only 100 clones that showed specificity

for α-syn fibrils and not monomers, which were then selected for further characterization. Isotyping of the

antibodies secreted by these clones revealed that only 33 of them were producing IgG antibodies, while the

remaining clones were producing antibodies either of the IgM type or a mixture. The clones producing IgM

were excluded and the hybridoma clones secreting IgG antibodies selected, passaged multiple times to

identify stable clones and subjected to single cell cloning to achieve monoclonality.

The specificity of the hybridoma clones for α-syn aggregates was assessed by dot blot (Figure 1).

Fourteen stable clones were found to be highly specific for α-syn aggregates and were passaged at least

three times prior to confirming their isotype subclass. Out of the 14 IgG-secreting clones, the best 6 clones

(based on the clones’ stability and their binding specificity towards α-syn aggregates) were selected for

extensive characterization, which were later named based on their preference to either early α-syn oligomers

and mature amyloid fibrils Syn-Os” or only to mature amyloid fibrils Syn-Fs” namely Syn-F1 (IgG1), Syn-

F2 (IgG2a), Syn-O1 (IgG2b), Syn-O2 (IgG1), Syn-O3 (IgG1) and Syn-O4 (IgG1).

Fig. 1. Representative image for the screening of hybridoma clones by dot blot.

50 ng of fibrils (F) and monomers (M) of α-syn were spotted onto a nitrocellulose membrane. The membranes were then probed with hybridoma culture supernatant (1:1 diluted in PBS) or control generic mAb Syn-1.

  50  

The selected mAbs only recognize fibrils of the full-length α-syn

In order to further evaluate the specificity of the selected antibodies for α-syn fibrils we performed an

inhibition ELISA and dot blotting. Inhibition ELISA was used to characterize the selectivity and affinity of

the six mAbs for α-syn fibrils versus monomers. According to the results, all six antibodies recognized

selectively α-syn fibrils compared to monomers (Figure 2A-F), whereas the control antibody Syn-1 detected

both α-syn fibrils and monomers (Figure 2G).

Fig. 2. Specificity of the Abs to α-syn fibrils. Inhibition ELISA using Syn-F1, Syn-F2, Syn-O1, Syn-O2, Syn-O3, Syn-O4 (A-F) and control mAb Syn-1 (G).

Our mAbs were selective for fibrils compared to the monomers whereas Syn-1 showed equal preference for both monomers and fibrils. (H) Dot blot to confirm the specificity of our mAbs to α-syn fibrils. 5 µg/ml of either α-syn fibrils or monomers were preabsorbed with 50 ng/ml of our mAbs or 200 ng/ml of control antibodies (Syn-1 or 211) for 2 h at RT. The mixture was added to nitrocellulose membrane coated (50 ng/spot) of α-syn fibrils (F) or monomers (M) and then incubated for 30 min at RT.

  51  

To confirm further the specificity of our antibodies for α-syn fibrils, we assessed their ability to detect

monomeric and fibrillar α-syn after preabsorption with α-syn fibrils or monomers. As expected,

preabsorption of the antibodies with α-syn fibrils completely abolished their ability to detect α-syn fibrils

(Figure 2H) whereas, preabsorption of the antibodies with α-syn monomers had no effect which was

comparable to the signal produced by the non-absorbed antibodies. In contrast, preabsorption of the control

antibodies (Syn-1 and 211) with either form of α-syn, hindered their ability to detect both monomeric and

fibrillar α-syn (Figure 2H). These results strongly suggest that the new mAbs generated recognize epitopes

that are exclusively present in α-syn fibrils.

Interestingly, by employing various fragments of α-syn, including the C-terminally truncated α-syn

(1-122), the central hydrophobic region of α-syn NAC (61-95) or the fragment NAC (61-78), we show that

all the newly generated mAbs only detect the fibrils generated by the full-length protein (Figure 3A). The

control antibodies (Syn-1 and 5C2) not only detected fibrils and monomers of full-length α-syn (1-140),

but also truncated α-syn (1-122) and the NAC (61-95) fragment.

To investigate further the selectivity of the six selected mAbs, we assessed their cross-reactivity

with monomeric or fibrillar forms of β- and γ-syn. As shown in Figure 3B, the antibodies did not cross-

react with either the monomers or the fibrils generated from β- or γ-syn. Similarly, the antibodies did not

detect the fibrillar forms of other amyloidogenic proteins including Aβ, Tau40, IAPP or ABri (Figure 3C).

These results further confirm the selectivity of the newly generated mAbs for α-syn fibrils.

  52  

Fig. 3. Testing the cross reactivity of our conformation-specific mAbs by dot blot.

50 ng of fibrils (F) ormonomers (M) fromfull length α-syn and fragments, α-syn (1-122), NAC (61-95) and NAC (61-78) (A),α-, β- and γ-syn (B) andα-syn, Tau40, Aβ42, IAPP and ABri (C)were spotted onto a nitrocellulosemembrane. Themembraneswere probed with ourmAbs or control antibodies as appropriate. Syn-1 for α-syn, anti-β-synuclein for β-syn and C-20 for γ-syn.5E2 for Tau40, 82E1 for Aβ, R10/99 for IAPP and rabbit anti-ABri antiserum for ABri. RLU/sec=relative light units/sec.

Next, we investigated the affinity of the generated mAbs for the fibrils versus that for the monomers

by their IC50 ELISA value, defined as the concentration of the antigen (α-syn monomers or fibrils) required

for inhibiting half of the maximum ELISA signal. Five of the six antibodies assessed exhibited an IC50

value for α-syn fibrils in pM range, while their corresponding IC50 values for α-syn monomers were in the

µM range (see Table 1). Overall, Syn-O2 showed the strongest affinity (~27000 fold) for α-syn fibrils

compared to monomers followed by Syn-F1 (~16000 fold) and Syn-O1 (~12000 fold) (see Table 1). Syn-

F2 showed the lowest affinity for fibrils compared to monomers as its IC50 values for α-syn fibrils and

monomers were 1.1 nM and 3.7 µM, respectively. Whereas, Syn-1 antibody that was employed as a

  53  

reference exhibited comparable affinities for α-syn monomers and fibrils with IC50 values for fibrils and

monomers being 2.1 nM and 1.0 nM, respectively (Table 1).

In order to assess the binding kinetics of the raised antibodies for α-syn fibrils, surface plasmon

resonance (SPR) binding studies were carried out. All six antibodies exhibited high affinity for α-syn fibrils

(Figure 4). Indeed, in accordance with the IC50 ELISA values obtained above, the affinity for α-syn fibrils

for the antibodies Syn-F1 and Syn-O1, -O2, -O3 and -O4 was in the pM range and for Syn-F2 in the nM

range, with Syn-O2 showing the highest (96 pM) and Syn-F2 showing the lowest (2.6 nM) affinity for α-

syn fibrils (see Table 1). Interestingly, the reference antibody 211 showed a KD of 2.5 nM, which is

comparable to the lowest affinity observed for Syn-F2 antibody (see Table 1). This apparent affinity is most

likely avidity and proper affinity assessments need testing with the Fab fragments of the antibodies.

Fig. 4. SPR sensogram showing the binding kinetics of our mAbs and control antibody 211 to α-syn fibrils. Binding of our mAbs was determined by surface plasmon resonance using BIAcore X biosensor.

The binding curve in each sensogram corresponds to different concentrations of the mAbs. The response unit (RU) was calculated for nonspecific binding relative to a blank flow cell surface.

  54  

The selected mAbs do not recognize the linear sequence of α-syn

To further assess whether the generated antibodies detect linear or conformational epitopes, we

evaluated their binding against an overlapping peptide library composed of synthetic 14-mer peptides,

corresponding to amino acid residues 1-14, 8-21, etc, and spanning the entire α-syn sequence (see Table 2).

As shown in Figure 5A, with the exception of peptide No.19, corresponding to the α-syn C-terminal

fragment 127-140, the generated mAbs did not detect any other α-syn peptides from the peptide library.

Nevertheless, the detection of α-syn (127-140) peptide was extremely weaker compared to their signal for

the detection of α-syn fibrils (Figure 5A). Moreover, the commercially available α-syn antibodies Syn-1

(recognizes amino acid residues 91-99 of α-syn) [41], N19 (recognizes the N-terminus of α-syn, i.e. amino

acid residues 1-60), FL-140 (recognizes the full length of α-syn), 3B6 (recognizes the amino acid residues

119-140 of α-syn) and 5C2 (recognizes the amino acid residues 61-95 of α-syn) were also employed as

controls in this experiment. All the control antibodies detected α-syn fibrils, but unlike the generated mAbs,

they also strongly detected the peptides that contained part of their epitopes (Figure 5A).

The ELISA results also revealed interesting information regarding the exposed part of the epitope

in the fibrils. Moreover, the blots of the ELISA plates exposed in X-ray film, shown in Figure 5B, illustrate

unambiguously the intensity of the ELISA signal, revealing that the control antibodies, which were raised

against the linear forms of α-syn detected both α-syn fibrils and the peptides corresponding to their epitopes

with comparable intensity. Conversely, the generated mAbs detected α-syn fibrils with much higher

intensity compared to the C-terminal peptide α-syn (127-140; peptide 19), which only detected a weak

signal (Figure 5B). This indicates that α-syn (127-140) may represent only a part of the epitope recognized

by the generated antibodies, or that it is partly exposed. Collectively, these results confirm further that Syn-

F1, -F2 and Syn-O1, -O2, -O3 and -O4 are conformation-specific antibodies, the epitope of which is partly

composed of the C-terminal region of α-syn.

  55  

Fig. 5. Epitope mapping using α-syn peptide library. (A) Pepscan by ELISA, mAbs syn-F1, Syn-F2, Syn-O1, Syn-O2, Syn-O3 and Syn-O4 showed a weak signal for the peptide number 19 (127-140).

The control antibodies reacted to the peptides corresponding to their epitopes. FL-140 recognizes the peptides number 9 (57-70), 16 (106-119) and 19 (127-140), N19 recognizes strongly peptide number 2 (8-21) andweak signal to peptides numbers 1 (1-14), 3 (15-28) and 6 (36-49), 3B6 recognizes strongly the peptide number 19 (127-140), Syn-1 recognizes peptide number 13 (85-98) and 5C2 recognizes the peptide number 12 (78-91). (B) Pepscan blots developed by exposing the plate onto an x-ray film after the ELISA signals were measured.

The conformation-specific mAbs exhibit different specificity for various α-syn species generated in

vitro

The specificity of the six antibodies was further evaluated by assessing their ability to detect early

aggregates of α-syn. For this purpose, α-syn was aggregated in vitro and samples were collected at different

time points over 48 h. The collected samples were spotted on a nitrocellulose membrane and probed with

the generated mAbs or the control antibody Syn-1. As shown in Figure 6A, B and D, the sample collected

at time 0 h contains monomers, and as expected none of the generated antibodies showed any

  56  

immunoreactivity. Interestingly, the antibodies Syn-O1, -O2, -O3 and –O4 exhibited immunoreactivity for

the samples collected at 2, 4, 6 and 16 h (Figure 6C), which presumably contain soluble low molecular

weight and high molecular weight oligomers (protofibrillar of α-syn (Figure 6A and D) defined based on

previous data [36, 42]). However, the immunoreactivity of Syn-O1, -O2, -O3 and –O4 antibodies was

stronger for the late aggregates present in the samples collected at 24, 36 and 48 h (Figure 6C). Interestingly,

the antibodies Syn-F1 and-F2 did not detect early aggregates, but they showed low immunoreactivity for

the 24 h sample and a much stronger signal for the samples generated thereafter, suggesting that these two

antibodies recognize more of the late aggregates of α-syn “mature fibrils”. The control antibody, Syn-1, on

the other hand, exhibited a uniform immunoreactivity for all the samples collected at different time points,

including the one collected at time 0 h (Figure 6C). Furthermore, EM confirmed that the samples collected

between 2-6 h contain only very small spherical oligomers and protofibrils as previously reported that only

monomers, dimers or small oligomers exist at the early stage of α-syn aggregation [43]. Whereas, the

samples at later time points contain mature amyloid fibrils (Figure 6D and Supplementary Figure S1).

In order to evaluate further the specificity of the generated antibodies for the samples collected at

various time points during the in vitro aggregation of α-syn, a sandwich ELISA was carried out employing

these mAbs for capturing the antigen. Consistent with the results of the dot blot, the antibodies Syn-O1, -

O2, -O3 and –O4 detected the α-syn species in the samples collected as early as 2 h and the level of detection

increased in a time-dependent fashion (Figure 6E). Furthermore, the antibodies Syn-F1 and Syn-F2 showed

a good level of detection for the samples collected at 24, 36 and 48 h that increased time-dependently

(Figure 6C). In contrast, the control antibody Syn-1 produced a good level of detection for all the collected

samples, indicating that Syn-1 exhibits an equal affinity for both monomers and aggregated species. These

data further support the notion that Syn-O1, -O2, -O3 and -O4 recognize both early oligomers and late

mature fibrils, whereas, Syn-F1 and -F2 prefer more of the mature amyloid fibrils.

Detection of high levels of presumable α-syn oligomers in brain lysates from DLB using the new

conformation-specific mAbs

To investigate whether the generated antibodies can detect, α-syn aggregates present in DLB brain, we

developed a sensitive ELISA employing the antibody Syn-O2 for capturing. Syn-O2 was selected based on

the fact that it has the highest affinity and recognizes both early oligomers and late α-syn fibrils (see Table

1 and Figure 6C and E). As shown in Figure 7A, the average levels of presumable α-syn oligomers were

significantly higher in DLB cases than the control (P=0.0012) and AD (P=0.0006) (Mann-Whitney U test),

with no significant difference observed between control and AD samples.

The homogenates of the frontal cortex samples of DLB, AD and control donors were further spotted onto

nitrocellulose membrane and probed with Syn-O2 to confirm the ELISA results. The immunoreactivity of

  57  

Syn-O2 was strong for DLB samples but relatively weak for AD and control brain samples (Figure 7B),

which is consistent with the oligomeric ELISA results.

Fig. 6. Time dependent aggregation assay.

Aggregation of α-syn was monitored by Th-S assay (A) and western blotting using FL-140 antibody (B). Samples collected at various time points (as indicated) were either used for dot blot by coating 50 ng/spot onto a nitrocellulose membrane and probing with our mAbs or Syn-1 antibody or sandwich ELISA by adding 5 nM of sample to ELISA plates coated with our mAbs or Syn-1 followed by detection with FL-140 antibody. By dot blot (C) and ELISA (E), mAbs Syn-O1, -O2, -O3 and –O4 exhibited weak signal at 2, 4, 6 and 16 h and strong signal for the late aggregates at 24, 36 and 48 whereas, the antibodies Syn-F1 and-F2 showed strong signal only for the aggregates generated at 24, 36 and 48 h. Electronmicrographs of samples collected at various time points as indicated during α-syn aggregation confirmed that the samples collected between 2-6 h contain only very small spherical oligomers and protofibrils, whereas, the samples at later time points contain mainly mature amyloid fibrils (D).

  58  

Fig. 7. Detection ofα-syn oligomers in human brain homogenates.

(A) Oligomeric ELISA using Syn-O2. The concentration of α-syn oligomers (pg/ml) in the brain homogenates, from control (solid circles), DLB (solid squares) and AD patients (solid triangles), was measured using Syn-O2-ELISA. There was a significant difference observed in the levels of α-syn oligomers between DLB and control (p b 0.0001) and also between DLB and AD (p b 0.0001) (one-way ANOVA with Dunnett post-hoc test). (B) Dot blot using Syn-O2. Brain extracts (0.5 µg of total protein) samples from DLB, AD and control were spotted onto nitrocellulose membrane, and then probed with Syn-O2.

Immunohistochemical analysis of human brain tissue Five laboratories evaluated the immunoreactivity of the six novel mAbs in postmortem human brain tisssue

samples in an independent manner.

Laboratory 1: In the SN of PD and DLB patients, a diversity of histopathological features were detected

with the six mAbs, including intracytoplasmic LBs and LNs, synaptic-like structures (<0.5 µm) and diffuse

intracytoplasmic immunostaining (Figure 8A). No immunoreactivity was observed in the SN of age-

matched non-demented controls with any of the novel mAbs (Figure 8A). Syn-O1-4 revealed more

synaptic-like immunostaining than Syn-F1 and Syn-F2 in both the PD and DLB cases. Both Syn-O2 and

Syn-O4 recognized small and long thin LNs in both DLB and PD cases. With the Syn-O1 antibody, few

immunostained-beaded LNs were observed in some controls in addition to synaptic immunoreactivity.

Interestingly, Syn-O1 and Syn-O3 revealed more LNs and synaptic immunoreactivity in the SN than the

commercial antibodies KM51 or Syn-1. Moreover, neurons with diffuse intracytoplasmic staining were

observed with the Syn-O1-4, whereas KM51 and Syn-1 stained mainly the larger intracytoplasmic LBs. In

addition, Syn-O1-4 immunoreactive LNs were observed in all PD and DLB donors, whereas mainly small

or long swollen/bulgy LNs were detected with the KM-51 and Syn-1 (Figure 8A). The differences between

the six mAbs were similar for PD and DLB cases. Similar immunoreactive structures were detected with

  59  

our mAbs in the entorhinal cortex and hippocampus of PD and DLB cases, including cortical LBs

particularly in the deeper layers and a dense meshwork of LNs in the superficial layers of the entorhinal

cortex (Figure 8B), whereas synaptic-like staining was observed in the Syn-O1-4 antibodies in the

hippocampus and entorhinal cortex (mainly superficial layers) of controls (Figure 8C). Syn-O1 and Syn-

O2 antibodies stained particularly smaller synaptic-like structures that were found within the total thickness

of the cortical grey matter. This synaptic pattern was not as well detected by Syn-F1 and -F2, with Syn-F1

though showing higher immunoreactivity than Syn-F2 to all detected structures.

Fig. 8. Representative photomicrographs of immunoreactivity with mAbs Syn-Os and Syn-Fs and commercial generic mAbs KM51 and Syn-1.

  60  

(A) Immunostaining in 10 µm-thick paraffin sections of post-mortem substantia nigra tissue of a non-demented control (female, 89 year), PD patient (male, 83 year; Braak PD stage 6) and DLB patient (female, 81 years; Braak PD stage 6, limbic dominant). No immunoreactivity with the mAbs was observed in the control case. In the PD and DLB cases, a diversity of histopathological features was detected with our novel conformation-specific antibodies, including LBs (arrowheads), LNs (*thin neurites;+bulgy neurite), diffuse intracellular staining, and synaptic-like immunostaining. The antibodies Syn-F1 and Syn-F2 and KM51 revealed predominantly LBs (arrowheads) and LNs (*), while synaptic-like immunostaining was less pronounced. Syn-1 stained LBs (arrowheads) andmany LNs (*) in the SN of the PD and DLB patients. Scale bar=50 µm. (B) Immunoreactivity withmAbs Syn-Os and Syn-Fs in six-adjacent 10 µmimmunostained paraffin sections of post-mortem entorhinal cortex and hippocampus of a DLB patient (male, 74 years). Comparison of the antibodies shows that Syn-O1 and Syn-O2 revealed the most synaptic-like staining in superficial layers of the entorhinal cortex. Both Syn-O2 and Syn-O4 provided strong immunoreactivity for small micro-aggregates and thin LNs. Syn-F1 and Syn-F2 revealed predominantly LBs and LNs,while synaptic-like stainingwas less pronounced. Syn-F1 hadmore affinity than Syn-F2. LBs and thick LNs aremainly seen in the deep layers of the entorhinal cortex. In the hippocampal CA1 area, LBs or small LNs were the main forms of α-syn immunoreactivity. Scale bar=50 µm. (C) Representative photomicrographs of immunoreactivity with mAbs Syn-Os and Syn-Fs in the entorhinal cortex and CA1 layer of the hippocampus of a non-demented control (male, 81 year). Synaptic-like immunoreactivitywas observed with Syn-O1-4 in the superficial layer, but not in deep layer of the entorhinal cortex and hippocampus. Synaptic-like staining was not observed with Syn-F1 and Syn-F2. Bar=50 µm, magnification 400x.

  61  

Laboratory 2: Pretreatment with proteinase K or autoclaving produced the best immunostaining results

revealing the dense meshwork of LNs and fine threads in DLB patients (Figure 9).

Fig. 9. Immunohistochemistry using mAbs Syn-Os and Syn-Fs with no pre-treatment or with three antigen retrieval methods (autoclave at 120 °C for 10 min in citrate buffer; 98 %formic acid for 15 min; 20 µg/ml of proteinase K for 5 min) in paraffin embedded tissue from entorhinal cortex of a DLB patient.

  62  

Parallel sectionswere cut in 6 µm- thick, deparaffinised, rehydrated followed epitope unmasking, all antibodies were diluted 1:5000 and incubated overnight at 4 °C. For detection, Histostain SP kit (Zymed, San Francisco, CA) was used with Romulin AEC chromogen (Biocare Medical,Walnut Creek, CA). All conditions identified the pathological neurites in the superficial layers but both autoclaving and proteinase K treatmentwere superior to identify the neuritic structures and also eliminated the diffuse staining of neuropil. Syn-F1 performed superior to Syn-F2whereas Syn-Os produced equal staining pattern. Scale bars=20 µm, magnification × 400.

Laboratory 3: Immunostaining of the anterior cingulate cortex in PD patients revealed that all six mAbs

labelled a large number of LBs predominantly in layer V neurons, sparse LNs and abundant synaptic-like

profiles, similar to the anti-phospho-S129 α-syn antibody (see Figure 10). In MSA, GCIs and a few LNs

were detected with all mAbs in the putamen of the case examined (Figure 10). LBs inclusion pathology

was not observed in these severely affected sections. The antibody against phospho-S129 α-syn identified

a similar number of GCIs, but did not show any synaptic α-syn immunoreactivity. Syn-F1 and Syn-F2

exhibited a staining pattern comparable to the pattern produced by the anti-phosphoS129 α-syn antibody.

Furthermore, similar to the anti-phospho-S129 α-syn, Syn-F1 and Syn-F2 demonstrated a weak

immunoreactivity for synaptic α-syn in all cases, including AD and controls (Figure 10). It is noteworthy

that synaptic α-syn immunoreactivity was prevalent with Syn-O1-4 mAbs, and particularly in AD cases

where neuronal loss was still minimal (Figure 10). In such cases there appeared to be an increase in both

neuronal density and synaptic material (Figure 10), potentially as tissue structures are initially affected by

the disease. Immunoreactivity was not observed in the white matter with any of the antibodies (see

Supplementary Figure S2).

  63  

Fig. 10. Immunostaining with mAbs Syn-Os, Syn-Fs, Syn-1 and anti-phospho S129, in the anterior

cingulate cortex from a case with PD, in the putamen from a case with MSA, in the anterior. or cingulate

cortex from a case with AD and in the anterior cingulate cortex from an aged, neurological control case.

All antibodies labelled LBs in PD, and glial cytoplasmic inclusions in MSA. When present, synaptic staining is most evident with Syn-Os. In AD where there is significant tissue shrinkage, there is an apparent increase in the density of both smaller neurons and synaptic immunoreactivity with the Syn-Os mAbs. Scale bar = 20 µm.

Laboratory 4: Double immunofluorescence staining with Syn-F2 or Syn-O4 and anti-Syn (MJFR1)

antibodies in the PD locus coeruleus revealed a high degree of colocalisation. Syn-F2 stained typical LBs

(Figure 11A1) and synaptic inclusions (Figure 11A2) in PD but not in a normal control (Figure 11A3). Syn-

O4 displayed immunoreactivity for LNs, (Figure 11A4), LBs (Figure 11A5) and small synaptic inclusions

(Figure 11A4 and 5) in PD but not in normal control (Figure 11A6).

  64  

Laboratory 5: LBs biochemically isolated from brain tissue were first identified by immunoreactivity for

the generic α-syn antibody, then scanned for the colocalization and staining patterns for each of our

conformation-specific antibody. All LBs, positive for generic sheep anti-α-syn antibody staining were also

positive for our mAbs. However, the staining using our mAbs was located in more peripheral regions of

LBs (Figure 11B). While the Syn-O1-O4 stained LBs with similar intensity to generic α-syn antibody

(Figure 11B), the Syn-F1 and Syn-F2 appeared weaker (Figure 11B).

Fig. 11. Confocal images of double-labeledα-syn-immunoreactive pathologies identified using conformation-specific mAbs and generic anti-α-syn antibodies.

(A) Representative confocal microscopic images of double immunofluorescent staining for Syn-F2, Syn-O4 (green) and the generic anti-α-syn antibody (MJFR1, red) antibodies in PD brain sections and normal control. Syn-F2 and Syn-O4 immunoreactivity for LBs and LNs co-localized to a high degree with MJFR1 signals in the PD brains sections. Syn-F2 stained typical LBs (1) and punctate inclusions (2) in the amygdala of the PD brain tissue but not in normal control (3). Syn-O4 displayed immunoreactivity for LNs (4), LBs (4) and small aggregates (5) in the locus ceruleus of the PD tissue but not in normal control (6). Scale bars=20 µm. (B) Colocalization of sheep anti-α-syn antibody (red) and the conformation-specificmAbs (green) in isolated LBs fromDLB brain tissue. All LBs that were positive for generic sheep anti-α-syn antibody staining were also positive for the conformation-specific mAbs. Syn-O1-O4 stained LBs with similar intensity to generic α-syn antibody (B, top 4 rows), while the Syn-F1 and Syn-F2 were weaker (B, bottom 2 rows). Scale bars= 10 µm.

  65  

Discussion

Several antibodies against α-syn have been developed since it was first detected as the major component of

LBs and GCIs [2, 28]. Indeed, the majority of α-syn antibodies have been developed against certain

fragments of α-syn [3, 44-47] or full length protein [48] and few raised against modified (i.e.in vitro

oxidized/nitrated α-syn) [49, 50], aggregated (under acidic or high salt conditions) [51, 52] or oligomerized

(induced by4-hydroxy-2-nonenal) α-syn [53] or purified LBs [4]. To enable the detection of

underestimated, or new pathological features, it is essential to develop α-syn antibodies, which recognize

specific α-syn conformations.

In order to generate mAbs that specifically target α-syn pathology, mice were immunized with in

vitro generated α-syn fibrils. Following the selection of the stable clones, determination of the antibody

isotype revealed that most of the hybridomas produced IgM antibodies rather than IgG. Similar findings

have been reported previously, indicating that the selection of conformation-specific monoclonal IgG-

producing hybridomas is difficult even by employing various immunization strategies [51, 52]. However,

14 hybridoma clones of IgG isotype were selected from 1100 clones that showed specificity towards α-syn

fibrils, six of which were selected for further characterization. The antibodies produced by these six clones

were designated Syn-F1, Syn-F2, Syn-O1, Syn-O2, Syn-O3 and Syn-O4 and characterized extensively. The

specificity of the generated mAbs was assessed by an inhibition ELISA and the results showed that the

mAbs were highly selective for α-syn fibrils. This finding was further confirmed by preabsorption of the

mAbs with either α-syn fibrils or monomer, proving their specificity for α-syn fibrils.

The affinity of the generated mAbs for the fibrils was determined by the IC50 values of the inhibition

ELISA. With the exception of the Syn-F2, which showed the lowest affinity for fibrils, the rest of the mAbs

exhibited IC50 values for the fibrils in pM range, whereas their corresponding IC50 values for α-syn

monomers were in the µM range. Interestingly, the IC50 values obtained for the six mAbs were much higher

compared to previously reported affinities of other mAbs directed for α-syn oligomers/ protofibrils [53].

The affinity of the generated antibodies was further confirmed by SPR analysis. Five of the generated mAbs

showed affinity for α-syn fibrils in pM range, with only Syn-F2 exhibiting affinity in the nM range. The

concentration of the fibrils was taken equivalent to the starting monomer concentration in the fresh sample.

This may not reflect the actual concentration of the fibrils per se which implies that the affinity of the mAbs

may be much higher to what we have observed. Moreover, the binding to the fibrils is most likely an avidity

effect and proper affinity assessment needs to be done using the Fab fragments.

The selectivity of the mAbs was further evaluated by assessing their cross-reactivity with

monomeric or fibrillar forms of β- or γ-syn, as well as other amyloidogenic proteins and peptides such as

Aβ, Tau40, IAPP or ABri. Surprisingly, none of the mAbs cross-reacted with the fibrils of β- or γ-syn, or

other amyloidogenic protein and peptides, further confirming that the raised mAbs are highly specific only

  66  

for α-syn fibrils. These findings are noteworthy considering that many mAbs against a particular

conformational epitope are less selective. It has been reported that polyclonal serum produced by

immunization of rabbits with molecular mimics of soluble oligomers of Aβ reacts equally with soluble

oligomers from other amyloid protein and peptides [54]. Moreover, antibodies raised against Aβ42 fibrils

recognize a generic amyloid epitope, as they react equally well with amyloid fibrils derived from other

proteins of unrelated sequence, such as transthyretin, IAPP, β-microglobulin, and polyglutamine [55].

The conformational specificity of our newly generated mAbs was further verified by pepscan using

a peptide library spanning the entire α-syn sequence. All antibodies exhibited a very weak reactivity for the

C-terminal peptide spanning the α-syn sequence between amino acids 127 and 140. This finding is in

agreement with the dot blotting results showing that none of the generated mAbs could detect the fibrils

formed by the C-terminally truncated α-syn (1-122) or the NAC fragments. In contrast, the control

antibodies FL-140 and 3B6, which also have epitope in the C-terminal region, showed almost equal affinity

to the peptide (127-140) and to α-syn fibrils. Taken together, these results show that the generated mAbs

do not recognize the linear peptides covering the α-syn sequence, and the C-terminus of α-syn may be a

part of the complex conformational epitope of α-syn amyloid fibrils. Our pepscan results revealed that the

C-terminal region of α-syn is more likely part of the conformational epitope for all our antibodies. However,

based on the observation that Syn-Fs prefer more of the mature amyloid fibrils of α-syn, and Syn-Os

recognize both early α-syn oligomers as well as late amyloid fibrils, these results suggest that the antibodies

are unlikely recognize exactly the same epitope sequences. Various modified forms of aggregated α-syn

have been identified in the LBs isolated from PD and DLB brain [56-58]. Interestingly, our mAbs also

recognized the aggregated forms of nitrated and phosphorylated α-syn (data not shown).

The specificity of the generated antibodies was further evaluated by assessing their ability to detect

α-syn aggregates generated in vitro at various time points. The aggregation of α-syn in vitro is a nucleation

dependent process, characterized by an initial lag phase, followed by a growth phase known as “elongation”

[59-61]. During the lag phase, there is accumulation of oligomers, which are considered more toxic than

fibrils [1, 17-19, 62], and in due course the oligomers convert to fibrils. Therefore, we investigated whether

the generated mAbs could show a differential binding specificity to early oligomers or late aggregates

formed during the in vitro aggregation of α-syn. The mAbs Syn-O1, -O2, -O3 and -O4 recognized both

early “oligomers” and late aggregates “amyloid fibrils”, whereas Syn-F1 and-F2 showed a preference for

the late aggregates.

To investigate whether the Syn-O’s could also detect the α-syn oligomers present in brain, we

employed Syn-O2 in a sandwich based ELISA to detect α-syn oligomers in human brain homogenates from

DLB, AD and control cases. The ELISA using Syn-O2 as a capturing antibody detected significantly higher

levels of α-syn oligomers in DLB samples compared to AD and control samples. These data are consistent

  67  

with our previous report demonstrating a significant increase in the levels of α-syn oligomers in the brain

of DLB compared to AD and controls [18].

The ability of the generated conformation-specific mAbs to detect α-syn pathology was further

explored by immunohistochemistry experiments using human PD, DLB, AD, MSA and control brain

sections. The mAbs labeled various histopathological features including LBs, LNs, synaptic pathology and

GCIs. Noteworthy, the Syn-Os were able to detect synaptic staining and LNs in the brain sections of PD

and DLB patients, whereas the Syn-Fs and commercially available generic antibodies (KM51 and Syn-1)

did not visualize these structures under the same conditions. In controls, no immunostaining was observed

in the substantia nigra with the Syn-Os and Syn-Fs, whereas synaptic staining was seen in the superficial

layers of the entorhinal cortex only with the Syn-Os. The synaptic staining with Syn-Os may suggest that

these antibodies recognize oligomeric species in normal brain which are not detected by Syn-Fs further

supporting the notion that the Syn-Fs recognize more specifically the late aggregates. The synaptic staining

observed is consistent with recent data showing multimeric complexes of α-syn are important

physiologically at synapses to attenuate synaptic vesicle recycling [63-66]. In PD, the mAbs labeled similar

structures in the brain sections to that observed with anti-phospho-S129 α-syn antibody, although a larger

proportion of large LBs were detected with the newly generated mAbs compared to the generic pan antibody

Syn-1. More inclusions were also identified with Syn-F2 and Syn-O4, than with Syn-F1. In MSA brain

sections, all antibodies labeled a comparable number of GCIs. A major advantage of the new conformation-

specific mAbs is that small micro-aggregates and very thin neurites in PD and DLB patients can be

recognized, which may contribute to a better understanding of the relevance of these small pathological

structures. Its noteworthy that the FILA-1 antibody, specific for aggregated α-syn was found to be specific

for the filamentous by more than 103-fold compared to the monomers but bound only poorly to isolated

LBs or DLB brain sections investigated by immunohistochemistry [67]. Comparison of antigen retrieval

methods showed that our mAbs stained pathological neurites in all conditions, allowing combinations with

other mAbs for colocalization studies. Autoclaving and proteinase K treatment improved the staining, as

previously described by others (Kovacs et al. 2012). Overall, our novel conformation-specific mAbs

distinguish pathological features in the elderly with high sensitivity and may serve as tools to characterize

α-syn pathology in more details in the brain and peripheral tissues.

Conformation-specific antibodies such as Syn-Fs, and Syn-Os may have numerous important

applications. They may prove invaluable in extending our knowledge on molecular pathogenesis of

synucleinopathies, allowing close monitoring of the formation of the pathogenic species during α-syn

aggregation in vitro and in vivo. Furthermore, antibodies such as Syn-Os maybe useful as diagnostic tools

to quantify α-syn oligomers in blood and/or CSF as biomarkers for synucleinopathies [62, 68]. Finally,

conformation-specific antibodies may also be useful as passive vaccines as anti-α-syn therapy. It has been

  68  

demonstrated that treatment of mice with monoclonal antibodies ameliorates the neuropathological and

behavioral deficits observed in transgenicα-syn mice [30, 69, 70].

In conclusion, we have generated and characterized six novel conformation-specific antibodies

with high affinity for α-syn aggregates. These antibodies recognize a widespread and distinct pattern of α-

syn related neuropathology including LBs, LNs, synaptic and GCIs. It is therefore likely that the antibodies

described herein can detect widespread α-syn pathology and possibly distinguish the various

neuropathological nuances that can be of certain diagnostic value. Such antibodies may also serve as

therapeutic agents in the form of vaccines in the field of passive immunotherapy, slowing or even reversing

α-syn aggregation.

Supplementary Figure 1. S1

Electron micrographs of samples collected at various time points as indicated during α-syn aggregation confirmed that the samples collected between 2-6 h contain only very small spherical oligomers and protofibrils, whereas, the samples at later time points contain mature amyloid fibrils.

  69  

Supplementary Figure 2. S2

Immunostaining with novel mAbs and control antibodies Syn-1 and phospho-S129 α-syn in the white matter underlying the anterior cingulate cortex from a case with Parkinson’s disease (A), in the posterior limb of the internal capsule from a case with multiple system atrophy (B), in the white matter underlying the anterior cingulate cortex from a case with Alzheimer’s disease (C) and in the white matter underlying the anterior cingulate cortex from an aged, neurological control case (D). Immunoreactivity was not observed in the white matter with any of the antibodies. Scale bar = 100 µm.

  70  

References  

[1]   K.  E.  Paleologou,  G.  B.  Irvine,  and  O.  M.  El-­‐Agnaf,  "Alpha-­‐synuclein  aggregation  in  neurodegenerative  diseases  and  its  inhibition  as  a  potential  therapeutic  strategy,"  Biochem  Soc  Trans,  vol.  33,  no.  Pt  5,  pp.  1106-­‐10,  Nov  2005.  

[2]   M.  G.  Spillantini,  R.  A.  Crowther,  R.  Jakes,  N.  J.  Cairns,  P.  L.  Lantos,  and  M.  Goedert,  "Filamentous  alpha-­‐synuclein  inclusions  link  multiple  system  atrophy  with  Parkinson's  disease  and  dementia  with  Lewy  bodies,"  Neurosci  Lett,  vol.  251,  no.  3,  pp.  205-­‐8,  Jul  31  1998.  

[3]   M.  G.  Spillantini,  R.  A.  Crowther,  R.  Jakes,  M.  Hasegawa,  and  M.  Goedert,  "alpha-­‐Synuclein  in  filamentous  inclusions  of  Lewy  bodies  from  Parkinson's  disease  and  dementia  with  lewy  bodies,"  Proc  Natl  Acad  Sci  U  S  A,  vol.  95,  no.  11,  pp.  6469-­‐73,  May  26  1998.  

[4]   M.  Baba  et  al.,  "Aggregation  of  alpha-­‐synuclein  in  Lewy  bodies  of  sporadic  Parkinson's  disease  and  dementia  with  Lewy  bodies,"  Am  J  Pathol,  Research  Support,  Non-­‐U.S.  Gov't  

                           Research  Support,  U.S.  Gov't,  P.H.S.  vol.  152,  no.  4,  pp.  879-­‐84,  Apr  1998.  [5]   M.  H.  Polymeropoulos  et  al.,  "Mutation  in  the  alpha-­‐synuclein  gene  identified  in  families  

with  Parkinson's  disease,"  (in  eng),  Science,  vol.  276,  no.  5321,  pp.  2045-­‐7,  Jun  27  1997.  [6]   R.  Krüger  et  al.,  "Ala30Pro  mutation  in  the  gene  encoding  alpha-­‐synuclein  in  Parkinson's  

disease,"  (in  eng),  Nat  Genet,  vol.  18,  no.  2,  pp.  106-­‐8,  Feb  1998.  [7]   J.  J.  Zarranz  et  al.,  "The  new  mutation,  E46K,  of  alpha-­‐synuclein  causes  Parkinson  and  

Lewy  body  dementia,"  (in  eng),  Ann  Neurol,  vol.  55,  no.  2,  pp.  164-­‐73,  Feb  2004.  [8]   S.  Appel-­‐Cresswell  et  al.,  "Alpha-­‐synuclein  p.H50Q,  a  novel  pathogenic  mutation  for  

Parkinson's  disease,"  (in  eng),  Mov  Disord,  vol.  28,  no.  6,  pp.  811-­‐3,  Jun  2013.  [9]   S.  Lesage  et  al.,  "G51D  α-­‐synuclein  mutation  causes  a  novel  parkinsonian-­‐pyramidal  

syndrome,"  (in  eng),  Ann  Neurol,  vol.  73,  no.  4,  pp.  459-­‐71,  Apr  2013.  [10]   M.  C.  Chartier-­‐Harlin  et  al.,  "Alpha-­‐synuclein  locus  duplication  as  a  cause  of  familial  

Parkinson's  disease,"  (in  eng),  Lancet,  vol.  364,  no.  9440,  pp.  1167-­‐9,  Sep  25-­‐Oct  1  2004.  [11]   O.  A.  Ross  et  al.,  "Genomic  investigation  of  alpha-­‐synuclein  multiplication  and  

parkinsonism,"  Ann  Neurol,  vol.  63,  no.  6,  pp.  743-­‐50,  Jun  2008.  [12]   B.  Winner  et  al.,  "In  vivo  demonstration  that  alpha-­‐synuclein  oligomers  are  toxic,"  (in  

eng),  Proc  Natl  Acad  Sci  U  S  A,  vol.  108,  no.  10,  pp.  4194-­‐9,  Mar  2011.  [13]   E.  Rockenstein  et  al.,  "Accumulation  of  oligomer-­‐prone  alpha-­‐synuclein  exacerbates  

synaptic  and  neuronal  degeneration  in  vivo,"  Brain,  vol.  137,  no.  Pt  5,  pp.  1496-­‐513,  May  2014.  

[14]   P.  J.  Kahle  et  al.,  "Subcellular  localization  of  wild-­‐type  and  Parkinson's  disease-­‐associated  mutant  alpha  -­‐synuclein  in  human  and  transgenic  mouse  brain,"  J  Neurosci,  vol.  20,  no.  17,  pp.  6365-­‐73,  Sep  1  2000.  

[15]   E.  Masliah  et  al.,  "Dopaminergic  loss  and  inclusion  body  formation  in  alpha-­‐synuclein  mice:  implications  for  neurodegenerative  disorders,"  (in  eng),  Science,  vol.  287,  no.  5456,  pp.  1265-­‐9,  Feb  2000.  

[16]   F.  L.  Martin,  S.  J.  Williamson,  K.  E.  Paleologou,  D.  Allsop,  and  O.  M.  El-­‐Agnaf,  "Alpha-­‐synuclein  and  the  pathogenesis  of  Parkinson's  disease,"  Protein  Pept  Lett,  vol.  11,  no.  3,  pp.  229-­‐37,  Jun  2004.  

  71  

[17]   T.  Tokuda  et  al.,  "Detection  of  elevated  levels  of  α-­‐synuclein  oligomers  in  CSF  from  patients  with  Parkinson  disease,"  (in  eng),  Neurology,  vol.  75,  no.  20,  pp.  1766-­‐72,  Nov  2010.  

[18]   K.  E.  Paleologou  et  al.,  "Detection  of  elevated  levels  of  soluble  alpha-­‐synuclein  oligomers  in  post-­‐mortem  brain  extracts  from  patients  with  dementia  with  Lewy  bodies,"  (in  eng),  Brain,  vol.  132,  no.  Pt  4,  pp.  1093-­‐101,  Apr  2009.  

[19]   O.  M.  El-­‐Agnaf  et  al.,  "Detection  of  oligomeric  forms  of  alpha-­‐synuclein  protein  in  human  plasma  as  a  potential  biomarker  for  Parkinson's  disease,"  (in  eng),  FASEB  J,  vol.  20,  no.  3,  pp.  419-­‐25,  Mar  2006.  

[20]   L.  Parnetti  et  al.,  "Differential  role  of  CSF  alpha-­‐synuclein  species,  tau,  and  Abeta42  in  Parkinson's  Disease,"  Front  Aging  Neurosci,  vol.  6,  p.  53,  2014.  

[21]   O.  Hansson  et  al.,  "Levels  of  cerebrospinal  fluid  alpha-­‐synuclein  oligomers  are  increased  in  Parkinson's  disease  with  dementia  and  dementia  with  Lewy  bodies  compared  to  Alzheimer's  disease,"  Alzheimers  Res  Ther,  vol.  6,  no.  3,  p.  25,  2014.  

[22]   R.  Sharon,  I.  Bar-­‐Joseph,  M.  P.  Frosch,  D.  M.  Walsh,  J.  A.  Hamilton,  and  D.  J.  Selkoe,  "The  formation  of  highly  soluble  oligomers  of  alpha-­‐synuclein  is  regulated  by  fatty  acids  and  enhanced  in  Parkinson's  disease,"  (in  eng),  Neuron,  vol.  37,  no.  4,  pp.  583-­‐95,  Feb  2003.  

[23]   J.  O.  Aasly  et  al.,  "Elevated  levels  of  cerebrospinal  fluid  alpha-­‐synuclein  oligomers  in  healthy  asymptomatic  LRRK2  mutation  carriers,"  Front  Aging  Neurosci,  vol.  6,  p.  248,  2014.  

[24]   L.  A.  Volpicelli-­‐Daley  et  al.,  "Exogenous  alpha-­‐synuclein  fibrils  induce  Lewy  body  pathology  leading  to  synaptic  dysfunction  and  neuron  death,"  Neuron,  vol.  72,  no.  1,  pp.  57-­‐71,  Oct  6  2011.  

[25]   K.  C.  Luk  et  al.,  "Pathological  alpha-­‐synuclein  transmission  initiates  Parkinson-­‐like  neurodegeneration  in  nontransgenic  mice,"  Science,  vol.  338,  no.  6109,  pp.  949-­‐53,  Nov  16  2012.  

[26]   K.  C.  Luk,  V.  M.  Kehm,  B.  Zhang,  P.  O'Brien,  J.  Q.  Trojanowski,  and  V.  M.  Lee,  "Intracerebral  inoculation  of  pathological  alpha-­‐synuclein  initiates  a  rapidly  progressive  neurodegenerative  alpha-­‐synucleinopathy  in  mice,"  (in  eng),  J  Exp  Med,  vol.  209,  no.  5,  pp.  975-­‐86,  May  7  2012.  

[27]   A.  N.  Sacino  et  al.,  "Intramuscular  injection  of  alpha-­‐synuclein  induces  CNS  alpha-­‐synuclein  pathology  and  a  rapid-­‐onset  motor  phenotype  in  transgenic  mice,"  Proc  Natl  Acad  Sci  U  S  A,  vol.  111,  no.  29,  pp.  10732-­‐7,  Jul  22  2014.  

[28]   M.  G.  Spillantini,  M.  L.  Schmidt,  V.  M.  Lee,  J.  Q.  Trojanowski,  R.  Jakes,  and  M.  Goedert,  "Alpha-­‐synuclein  in  Lewy  bodies,"  (in  eng),  Nature,  vol.  388,  no.  6645,  pp.  839-­‐40,  Aug  1997.  

[29]   E.  Croisier  et  al.,  "Comparative  study  of  commercially  available  anti-­‐alpha-­‐synuclein  antibodies,"  Neuropathol  Appl  Neurobiol,  Comparative  Study  

Research  Support,  Non-­‐U.S.  Gov't  vol.  32,  no.  3,  pp.  351-­‐6,  Jun  2006.  [30]   E.  Masliah  et  al.,  "Passive  immunization  reduces  behavioral  and  neuropathological  

deficits  in  an  alpha-­‐synuclein  transgenic  model  of  Lewy  body  disease,"  PLoS  One,  Research  Support,  N.I.H.,  Extramural  

Research  Support,  Non-­‐U.S.  Gov't  vol.  6,  no.  4,  p.  e19338,  2011.  

  72  

[31]   D.  Games  et  al.,  "Reducing  C-­‐terminal-­‐truncated  alpha-­‐synuclein  by  immunotherapy  attenuates  neurodegeneration  and  propagation  in  Parkinson's  disease-­‐like  models,"  J  Neurosci,  vol.  34,  no.  28,  pp.  9441-­‐54,  Jul  9  2014.  

[32]   E.  Masliah  et  al.,  "Effects  of  alpha-­‐synuclein  immunization  in  a  mouse  model  of  Parkinson's  disease.,"  (in  eng),  Neuron,  vol.  46,  no.  6,  pp.  857-­‐68,  Jun  2005.  

[33]   M.  T.  Ardah  et  al.,  "Structure  activity  relationship  of  phenolic  acid  inhibitors  of  alpha-­‐synuclein  fibril  formation  and  toxicity,"  Front  Aging  Neurosci,  vol.  6,  p.  197,  2014.  

[34]   O.  M.  El-­‐Agnaf  et  al.,  "Aggregates  from  mutant  and  wild-­‐type  alpha-­‐synuclein  proteins  and  NAC  peptide  induce  apoptotic  cell  death  in  human  neuroblastoma  cells  by  formation  of  beta-­‐sheet  and  amyloid-­‐like  filaments,"  (in  eng),  FEBS  Lett,  vol.  440,  no.  1-­‐2,  pp.  71-­‐5,  Nov  1998.  

[35]   S.  T.  O'Dowd  et  al.,  "The  ELISA-­‐measured  increase  in  cerebrospinal  fluid  tau  that  discriminates  Alzheimer's  disease  from  other  neurodegenerative  disorders  is  not  attributable  to  differential  recognition  of  tau  assembly  forms,"  J  Alzheimers  Dis,  vol.  33,  no.  4,  pp.  923-­‐8,  2013.  

[36]   O.  M.  El-­‐Agnaf  et  al.,  "Effect  of  the  disulfide  bridge  and  the  C-­‐terminal  extension  on  the  oligomerization  of  the  amyloid  peptide  ABri  implicated  in  familial  British  dementia,"  (in  eng),  Biochemistry,  vol.  40,  no.  12,  pp.  3449-­‐57,  Mar  2001.  

[37]   O.  M.  El-­‐Agnaf,  H.  Goodwin,  J.  M.  Sheridan,  E.  R.  Frears,  and  B.  M.  Austen,  B.  M.  Dunn,  Ed.  Improved  solid-­‐phase  syntheses  of  amyloid  proteins  associated  with  neurodegenerative  diseases,  1  ed.  Bentham  Science  Publishers  B.  V,  2000,  p.  8.  

[38]   M.  T.  Ardah  et  al.,  "Ginsenoside  Rb1  inhibits  fibrillation  and  toxicity  of  alpha-­‐synuclein  and  disaggregates  preformed  fibrils,"  Neurobiol  Dis,  vol.  74,  pp.  89-­‐101,  Feb  2015.  

[39]   W.  P.  Gai,  H.  X.  Yuan,  X.  Q.  Li,  J.  T.  Power,  P.  C.  Blumbergs,  and  P.  H.  Jensen,  "In  situ  and  in  vitro  study  of  colocalization  and  segregation  of  alpha-­‐synuclein,  ubiquitin,  and  lipids  in  Lewy  bodies,"  Exp  Neurol,  vol.  166,  no.  2,  pp.  324-­‐33,  Dec  2000.  

[40]   W.  P.  Gai,  J.  H.  Power,  P.  C.  Blumbergs,  J.  G.  Culvenor,  and  P.  H.  Jensen,  "Alpha-­‐synuclein  immunoisolation  of  glial  inclusions  from  multiple  system  atrophy  brain  tissue  reveals  multiprotein  components,"  J  Neurochem,  vol.  73,  no.  5,  pp.  2093-­‐100,  Nov  1999.  

[41]   R.  J.  Perrin  et  al.,  "Epitope  mapping  and  specificity  of  the  anti-­‐alpha-­‐synuclein  monoclonal  antibody  Syn-­‐1  in  mouse  brain  and  cultured  cell  lines,"  (in  eng),  Neurosci  Lett,  vol.  349,  no.  2,  pp.  133-­‐5,  Oct  2003.  

[42]   D.  M.  Walsh,  A.  Lomakin,  G.  B.  Benedek,  M.  M.  Condron,  and  D.  B.  Teplow,  "Amyloid  beta-­‐protein  fibrillogenesis.  Detection  of  a  protofibrillar  intermediate,"  J  Biol  Chem,  vol.  272,  no.  35,  pp.  22364-­‐72,  Aug  29  1997.  

[43]   H.  Zhang,  A.  Griggs,  J.  C.  Rochet,  and  L.  A.  Stanciu,  "In  vitro  study  of  alpha-­‐synuclein  protofibrils  by  cryo-­‐EM  suggests  a  Cu(2+)-­‐dependent  aggregation  pathway,"  Biophys  J,  vol.  104,  no.  12,  pp.  2706-­‐13,  Jun  18  2013.  

[44]   D.  W.  Dickson  et  al.,  "Widespread  alterations  of  alpha-­‐synuclein  in  multiple  system  atrophy,"  (in  eng),  Am  J  Pathol,  vol.  155,  no.  4,  pp.  1241-­‐51,  Oct  1999.  

[45]   J.  G.  Culvenor  et  al.,  "Non-­‐Abeta  component  of  Alzheimer's  disease  amyloid  (NAC)  revisited.  NAC  and  alpha-­‐synuclein  are  not  associated  with  Abeta  amyloid,"  (in  eng),  Am  J  Pathol,  vol.  155,  no.  4,  pp.  1173-­‐81,  Oct  1999.  

  73  

[46]   B.  I.  Giasson  et  al.,  "A  panel  of  epitope-­‐specific  antibodies  detects  protein  domains  distributed  throughout  human  alpha-­‐synuclein  in  Lewy  bodies  of  Parkinson's  disease,"  J  Neurosci  Res,  Research  Support,  Non-­‐U.S.  Gov't  

Research  Support,  U.S.  Gov't,  P.H.S.  vol.  59,  no.  4,  pp.  528-­‐33,  Feb  15  2000.  [47]   R.  Jakes,  M.  G.  Spillantini,  and  M.  Goedert,  "Identification  of  two  distinct  synucleins  

from  human  brain,"  FEBS  Lett,  vol.  345,  no.  1,  pp.  27-­‐32,  May  23  1994.  [48]   S.  Yu  et  al.,  "Extensive  nuclear  localization  of  alpha-­‐synuclein  in  normal  rat  brain  

neurons  revealed  by  a  novel  monoclonal  antibody,"  (in  eng),  Neuroscience,  vol.  145,  no.  2,  pp.  539-­‐55,  Mar  2007.  

[49]   J.  E.  Duda,  B.  I.  Giasson,  M.  E.  Mabon,  V.  M.  Lee,  and  J.  Q.  Trojanowski,  "Novel  antibodies  to  synuclein  show  abundant  striatal  pathology  in  Lewy  body  diseases,"  Ann  Neurol,  Research  Support,  Non-­‐U.S.  Gov't  

Research  Support,  U.S.  Gov't,  P.H.S.  vol.  52,  no.  2,  pp.  205-­‐10,  Aug  2002.  [50]   E.  A.  Waxman,  J.  E.  Duda,  and  B.  I.  Giasson,  "Characterization  of  antibodies  that  

selectively  detect  alpha-­‐synuclein  in  pathological  inclusions,"  Acta  Neuropathol,  Research  Support,  N.I.H.,  Extramural  vol.  116,  no.  1,  pp.  37-­‐46,  Jul  2008.  

[51]   A.  L.  Mougenot  et  al.,  "Production  of  a  monoclonal  antibody,  against  human  alpha-­‐synuclein,  in  a  subpopulation  of  C57BL/6J  mice,  presenting  a  deletion  of  the  alpha-­‐synuclein  locus,"  J  Neurosci  Methods,  Research  Support,  Non-­‐U.S.  Gov't  vol.  192,  no.  2,  pp.  268-­‐76,  Oct  15  2010.  

[52]   G.  G.  Kovacs  et  al.,  "An  antibody  with  high  reactivity  for  disease-­‐associated  α-­‐synuclein  reveals  extensive  brain  pathology,"  (in  eng),  Acta  Neuropathol,  vol.  124,  no.  1,  pp.  37-­‐50,  Jul  2012.  

[53]   T.  Fagerqvist  et  al.,  "Monoclonal  antibodies  selective  for  alpha-­‐synuclein  oligomers/protofibrils  recognize  brain  pathology  in  Lewy  body  disorders  and  alpha-­‐synuclein  transgenic  mice  with  the  disease-­‐causing  A30P  mutation,"  J  Neurochem,  Research  Support,  Non-­‐U.S.  Gov't  vol.  126,  no.  1,  pp.  131-­‐44,  Jul  2013.  

[54]   R.  Kayed  et  al.,  "Common  structure  of  soluble  amyloid  oligomers  implies  common  mechanism  of  pathogenesis,"  (in  eng),  Science,  vol.  300,  no.  5618,  pp.  486-­‐9,  Apr  2003.  

[55]   B.  O'Nuallain  and  R.  Wetzel,  "Conformational  Abs  recognizing  a  generic  amyloid  fibril  epitope,"  Proc  Natl  Acad  Sci  U  S  A,  vol.  99,  no.  3,  pp.  1485-­‐90,  Feb  5  2002.  

[56]   H.  Fujiwara  et  al.,  "alpha-­‐Synuclein  is  phosphorylated  in  synucleinopathy  lesions,"  Nat  Cell  Biol,  vol.  4,  no.  2,  pp.  160-­‐4,  Feb  2002.  

[57]   B.  I.  Giasson  et  al.,  "Oxidative  damage  linked  to  neurodegeneration  by  selective  alpha-­‐synuclein  nitration  in  synucleinopathy  lesions,"  Science,  Research  Support,  Non-­‐U.S.  Gov't  

Research  Support,  U.S.  Gov't,  P.H.S.  vol.  290,  no.  5493,  pp.  985-­‐9,  Nov  3  2000.  [58]   J.  P.  Anderson  et  al.,  "Phosphorylation  of  Ser-­‐129  is  the  dominant  pathological  

modification  of  alpha-­‐synuclein  in  familial  and  sporadic  Lewy  body  disease,"  J  Biol  Chem,  vol.  281,  no.  40,  pp.  29739-­‐52,  Oct  6  2006.  

[59]   S.  J.  Wood,  J.  Wypych,  S.  Steavenson,  J.  C.  Louis,  M.  Citron,  and  A.  L.  Biere,  "alpha-­‐synuclein  fibrillogenesis  is  nucleation-­‐dependent.  Implications  for  the  pathogenesis  of  Parkinson's  disease,"  (in  eng),  J  Biol  Chem,  vol.  274,  no.  28,  pp.  19509-­‐12,  Jul  1999.  

  74  

[60]   N.  Cremades  et  al.,  "Direct  observation  of  the  interconversion  of  normal  and  toxic  forms  of  α-­‐synuclein,"  (in  eng),  Cell,  vol.  149,  no.  5,  pp.  1048-­‐59,  May  2012.  

[61]   O.  M.  El-­‐Agnaf,  G.  B.  Irvine,  and  D.  J.  Guthrie,  "Conformations  of  beta-­‐amyloid  in  solution,"  J  Neurochem,  vol.  68,  no.  1,  pp.  437-­‐9,  Jan  1997.  

[62]   O.  M.  El-­‐Agnaf,  D.  M.  Walsh,  and  D.  Allsop,  "Soluble  oligomers  for  the  diagnosis  of  neurodegenerative  diseases.,"  (in  eng),  Lancet  Neurol,  vol.  2,  no.  8,  pp.  461-­‐2,  Aug  2003.  

[63]   J.  Burre,  M.  Sharma,  and  T.  C.  Sudhof,  "alpha-­‐Synuclein  assembles  into  higher-­‐order  multimers  upon  membrane  binding  to  promote  SNARE  complex  formation,"  Proc  Natl  Acad  Sci  U  S  A,  vol.  111,  no.  40,  pp.  E4274-­‐83,  Oct  7  2014.  

[64]   L.  Wang,  U.  Das,  D.  A.  Scott,  Y.  Tang,  P.  J.  McLean,  and  S.  Roy,  "alpha-­‐synuclein  multimers  cluster  synaptic  vesicles  and  attenuate  recycling,"  Curr  Biol,  vol.  24,  no.  19,  pp.  2319-­‐26,  Oct  6  2014.  

[65]   Y.  Lai  et  al.,  "Nonaggregated  alpha-­‐synuclein  influences  SNARE-­‐dependent  vesicle  docking  via  membrane  binding,"  Biochemistry,  vol.  53,  no.  24,  pp.  3889-­‐96,  Jun  24  2014.  

[66]   B.  K.  Choi  et  al.,  "Large  alpha-­‐synuclein  oligomers  inhibit  neuronal  SNARE-­‐mediated  vesicle  docking,"  Proc  Natl  Acad  Sci  U  S  A,  vol.  110,  no.  10,  pp.  4087-­‐92,  Mar  5  2013.  

[67]   E.  Lindersson  et  al.,  "Proteasomal  inhibition  by  alpha-­‐synuclein  filaments  and  oligomers,"  J  Biol  Chem,  vol.  279,  no.  13,  pp.  12924-­‐34,  Mar  26  2004.  

[68]   L.  Parnetti  et  al.,  "Cerebrospinal  fluid  biomarkers  in  Parkinson  disease,"  Nat  Rev  Neurol,  vol.  9,  no.  3,  pp.  131-­‐40,  Mar  2013.  

[69]   H.  T.  Tran  et  al.,  "α-­‐Synuclein  Immunotherapy  Blocks  Uptake  and  Templated  Propagation  of  Misfolded  α-­‐Synuclein  and  Neurodegeneration,"  (in  eng),  Cell  Rep,  vol.  7,  no.  6,  pp.  2054-­‐65,  Jun  2014.  

[70]   E.  J.  Bae  et  al.,  "Antibody-­‐Aided  Clearance  of  Extracellular  α-­‐Synuclein  Prevents  Cell-­‐to-­‐Cell  Aggregate  Transmission,"  (in  eng),  J  Neurosci,  vol.  32,  no.  39,  pp.  13454-­‐69,  Sep  2012.  

     

 

 

 

 

 

   

  75    

!!

CHAPTER 3

Oligomeric and Phosphorylated Alpha-Synuclein as Potential CSF Biomarkers for Parkinson’s Disease

  76  

 CHAPTER 3 | OLIGOMERIC AND PHOSPHORYLATED ALPHA-SYNUCLEIN AS POTENTIAL CSF BIOMARKERS FOR PARKINSON’S DISEASE  

Nour K. Majbour, MSc1,2, Nishant N. Vaikath, MSc1,3, Karin D. van Dijk, PhD2, Mustafa T. Ardah, PhD1, Shiji Varghese, MSc1, Louise B. Vesterager, PhD4, Liliana P. Montezinho, PhD4, Stephen Poole, PhD5, Bared Safieh-Garabedian, PhD6, Takahiko Tokuda, MD, PhD7, Charlotte E. Teunissen, MD, PhD8, HenkW. Berendse, MD, PhD9, Wilma D.J. van de Berg, PhD2 and Omar MA El-Agnaf, PhD 10

1Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates

2Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU University Medical Centre, Amsterdam, The Netherlands

3Neural Plasticity and Repair Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, BMC A10, Lund University, Lund, Sweden 4Department of Neurodegeneration, H. Lundbeck A/S, 2500 Valby, Denmark 5Biotherapeutics Group, National Institute for Biological Standards and Control, Potters Bar, Herts, UK 6College of Medicine, Qatar University, Doha, Qatar 7Department of Neurology, Research Institute for Geriatrics, Kyoto Prefectural University of Medicine, Kyoto, 602-0841, Japan 8Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Neuroscience Campus Amsterdam, VU University Medical Centre, Amsterdam, The Netherlands 9Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Centre, Amsterdam, The Netherlands 10Neurological Disorders Center, Qatar Biomedical Research Institute, and College of Science and Engineering, Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, P.O. Box 5825 Doha, Qatar  Mol Neurodegener. 2016 Jan 19;11:7. doi: 10.1186/s13024-016-0072-9.

  77  

Abstract

Background: Despite decades of intensive research, to date, there is no accepted diagnosis for Parkinson’s

disease (PD) based on biochemical analysis of blood or CSF. However, neurodegeneration in the brains of

PD patients begins several years before the manifestation of the clinical symptoms, pointing to serious

flaw/limitations in this approach.

Results: To explore the potential use of alpha-synuclein (α-syn) species as candidate biomarkers for PD,

we generated specific antibodies directed against wide array of α-syn species, namely total-, oligomeric-

and phosphorylated-Ser129-α-syn (t-, o- and p-S129-α-syn). Next we sought to employ our antibodies to

develop highly specific ELISA assays to quantify α-syn species in biological samples. Finally we verified

the usefulness of our assays in CSF samples from 46 PD patients and 48 age-matched healthy controls. We

also assessed the discriminating power of combining multiple CSF α-syn species with classical Alzheimer's

disease biomarkers. The combination of CSF o-/t-α-syn, p-S129-α-syn and p-tau provided the best fitting

predictive model for discriminating PD patients from controls. Moreover, CSF o-α-syn levels correlated

significantly with the severity of PD motor symptoms (r = -0.37), whereas CSF t-α-syn correlated inversely

with MMSE scores (r = -0.46).

Conclusion: Our new ELISA assays can serve as research tools to address the unmet need for reliable CSF

biomarkers for PD and related disorders.

Keywords: Parkinson’s disease, alpha synuclein, cerebrospinal fluid biomarkers, amyloid oligomers,

biomarkers

Background

Parkinson’s disease (PD) is the second most common neurodegenerative disorder after Alzheimer’s disease.

PD is characterized by a large time-gap between the beginning of the neurodegenerative processes and the

onset of clinical manifestations [1]. However, its diagnosis is primarily based on motor-related clinical

criteria. The natural history of the disease includes an initial asymptomatic stage followed by a long pre-

motor phase; finally, when the classical motor symptoms appear, a minimum of 30% of nigral dopaminergic

neurons have already degenerated [2-5]. Therefore, there is an urgent need for the discovery of reliable

biomarkers for PD diagnosis and prognosis. Toward this aim, recent intensive investigations have attempted

to identify biomarker(s) to enable an early diagnosis of PD, preferably during the pre-motor phase. And

since molecular changes in the brain can be reflected in the CSF proteome, this makes CSF an ideal source

of reliable biomarkers [6]. Aggregated and hyper-phosphorylated forms of alpha-synuclein (α-syn) protein

at S129 are the major components of Lewy bodies (LBs) and Lewy neurites and constitute the hallmark

pathology in the brains of patients with PD and dementia with Lewy bodies [7]. Although the accumulation

  78  

of α-syn is central to the development of synucleinopathies, the precise pathophysiological mechanism

remains uncertain [8]. Of the different species of α-syn, intermediate oligomers/protofibrils are thought to

be the major neurotoxic species [9, 10]. Therefore, developing therapeutic and diagnostic strategies that

target these species is of paramount importance. Several retrospective studies have explored CSF α-syn as

a putative biomarker for PD. A clear trend of lower CSF t-α-syn levels or marginally higher p-S129-α-

syn/t-α-syn ratio in PD have been reported, although a large overlap between PD and control groups was

noted [11-17]. Clearly, from previous studies it is evident that t-α-syn levels alone cannot discriminate

individual patients with synucleinopathies from healthy individuals or those with other neurodegenerative

diseases. Soluble α-syn oligomers (o-α-syn) have been increasingly linked to synaptic and neuronal

degeneration [18] . The exact mechanism by which α-syn oligomers cause neuronal cell death is not fully

elucidated. It has been shown that soluble o-α-syn is elevated in brain homogenates from PD and DLB

cases compared with controls [19, 20]. Interestingly, we have previously reported that CSF o-α-syn levels

were significantly higher in PD patients compared with other neurological disorders [14, 16]. CSF o-α-syn

levels and o-α-syn/t-α-syn ratios were found to be substantially higher in patients with PD, including those

with mild and early-stage disease, compared with controls [14, 16, 21-23].

Considering the findings presented above, it’s crucial to adopt precise strategies/methods capable to

specifically quantify α-syn neurotoxic species in biological samples. In this context, we demonstrated the

development of specific, sensitive and reliable ELISA assays using our antibodies to assess different species

of α-syn (t-, o- and p-S129-α-syn) in different biological samples. The provision of such reliable

immunoassays is not only valuable for the discovery of ideal CSF biomarkers but also for a better

understanding of the underlying pathophysiological process in PD and related disorders. Next, to explore

the potential of our novel assays, we employed them to quantify different species of α-syn in CSF from a

cohort of PD patients and age-matched healthy controls (HC). Finally, we investigated the usefulness of

combining several biomarkers that reflect different pathogenic pathways, including AD classical

biomarkers (total-tau (t-tau), phosphorylated-tau (p-tau) and amyloid-beta-42 (Aβ-42)), as candidate CSF

biomarkers for PD and related disorders.

Methods

Generation of anti-α-syn antibodies:

Generation and purification of our mouse monoclonal antibodies were performed as previously described

[24]. Briefly, the antigens, recombinant human α-syn protein, α-syn fibrils [25] or synthetic

phosphopeptide-S129 (CYEMPSEEGY (α-syn aa 125 to 133; [26, 27]) were used to generate 11D12, Syn-

O2 and PS129 mouse monoclonal antibodies respectively. The antigens were repeatedly used to immunize

female BALB/C mice subcutaneously. Mice with appropriate plasma titers were euthanized and splenocytes

were extracted and fused with Sp2/0 myeloma cells and then seeded in HAT (hypoxantin, aminopterine,

  79  

and thymidine) selective medium to generate antibody-producing hybridomas according to standard

techniques. After 10 days, the medium was replaced with hypoxantin, thymidine medium and 5 days later

the culture supernatants were tested for secreted anti-α-syn antibodies using indirect ELISA. Several

monoclonal antibodies were produced, purified and thoroughly characterized, among which Syn-O2

(IgG1), 11D12 (IgG2a) and PS129 (IgG2a) were selected for this study. The isotypes were determined

using an isotyping kit (Sigma-Aldrich, USA) and the antibody purification was done using Protein G-

agarose (Sigma-Aldrich, USA) affinity chromatography.

Syn-140 is a sheep polyclonal antibody raised against full-length human α-syn as previously described [28,

29]. In brief, a polyclonal antiserum was raised against α-syn in sheep by intramuscular injection into four

sites of 500 µg recombinant α-syn protein emulsified in Freund's complete adjuvant. Boosts of 200 µg in

Freund's incomplete adjuvant were given intramuscularly into four sites at intervals after the primary

injection of 1 month, 2 months and 6 months. The sheep were bled 8 days after the final boost.

Dot Blot

Monomeric and aggregated forms of recombinant α-, β- and γ-syn or tau proteins and synthetic peptides

Aβ-42 [9, 30]; Islet Amyloid Polypeptide (IAPP) [31] ; or ABri [26] were used for testing the specificity of

Syn-O2. Each protein/peptide was spotted (50 ng) onto a nitrocellulose membrane and allowed to dry at

RT for 1 h. The membranes were then incubated for 1 h at RT with 5% skimmed milk in PBST (PBS

containing 0.05% Tween-20) with gentle agitation. The blocking solution was then removed, and the

membranes were incubated for 2 h at RT with the primary antibodies: Syn-O2; mouse anti-α-syn (Syn-1,

BD Bioscience); mouse anti-β-syn (8; Santa Cruz Biotechnology); goat anti-γ-syn (C-20, Santa Cruz

Biotechnology); mouse anti-tau (5E2, Millipore); mouse anti-amyloid beta (82E1, IBL) or mouse anti-

amylin (R10/99, Santa Cruz Biotechnology). The membranes were then washed three times with PBST and

incubated with secondary antibodies, goat anti-mouse HRP (1:20 K, Jackson Immunoresearch, PA, USA);

goat anti-rabbit (1:20 K, Jackson Immunoresearch, PA, USA) or chicken anti-goat HRP ( 1:30 K, Santa

Cruz Biotechnology) as appropriate for 1 h at RT. The membranes were then washed three times with PBST

and developed using SuperSignal West Pico Chemiluminescent Substrate (Pierce).

Western Blotting

Samples of human-, mouse- and rat-brain lysates (15 µg), recombinant α-, β- and γ-syn or human and mouse

p-S129-α-syn (50 ng) were mixed with loading buffer (250 mM Tris-HCl, pH 6.8, 30% glycerol, 0.02%

bromophenol blue) and then separated on 15% SDS-PAGE gels. The separated proteins were transferred to

0.45 µm nitrocellulose membranes (WhatmanGmbh-Germany) at 100 V for 45 min. The membranes were

boiled for 5 min in PBS and then blocked for 1 h with 5% non-fat milk prepared in PBST prior to incubation

with 10 ml of primary antibody (50 ng/ml) for 2 h at RT. The membranes were then washed 4 times with

PBST followed by incubation with the secondary antibody. After one hour, the membranes were washed 4

  80  

times with PBST, and immunoreactive bands were visualized using the SuperSignal West Pico

Chemiluminescent Substrate Kit (Pierce) according to the manufacturer’s instructions.

Preparation of the standard for the oligomeric ELISA

Expression and purification of recombinant human α-syn was performed as previously described [25]. α-

Syn oligomers were prepared by incubating fresh α-syn (25 µM) with Ginsenoside Rb1 (100 µM) in PBS

at 37°C for 5 days with continuous shaking at 800 rpm in a Thermomixer (Eppendorf), then α-syn oligomers

were characterized using electron microscopy as previously described [32]. Briefly, size exclusion

chromatography was carried out using an AKTA FPLC system (GE Healthcare, Sweden) and a Superdex

200 column at 4 °C, in order to separate soluble α-syn oligomers from monomers. This way, we guaranteed

that only pure soluble α-syn oligomers were separated and then confirmed by electron microscopy (Figure

4A) [32].

Development of ELISA for measuring t-α-syn in human CSF

A 384-well ELISA microplate (Nunc MaxiSorb, NUNC) was coated by overnight incubation at 4°C with

0.1 µg/ml Syn-140 (sheep anti-α-syn polyclonal antibody) in 200 mM NaHCO3, pH 9.6 (50 µl/well). The

plate was then washed with PBST and incubated with 100 µl/well of blocking buffer (PBST containing

2.5% gelatin) for 2 hours at 37°C. After washing, 50 µl of the CSF samples (thawed on ice and Tween-20

was added to a final concentration of 0.05%) was added to each well, and plates were incubated at 37°C for

2.5 hours. 11D12 (mouse anti-α-syn monoclonal antibody), diluted in blocking buffer at 1:5 K was added

to the appropriate wells, and incubated at 37°C for 2 hours. Next, the plate was washed and incubated for

2 hours at 37°C with 50 µl/well of species-appropriate secondary antibody (donkey anti-mouse IgG HRP,

Jackson ImmunoResearch) diluted in blocking buffer (1:20 K). After washing, the plate was incubated with

50 µl/well of an enhanced chemiluminescent substrate (SuperSignal ELISA Femto, Pierce Biotechnology,

Rockford, IL). The chemiluminescence, expressed in relative light units, was immediately measured using

VICTOR™ X3 multilabel plate reader (PerkinElmer). The standard curve for the ELISA assay was carried

out using serial dilutions of recombinant human α-syn in artificial CSF.

Development of ELISA for measuring p-S129-α-syn in human CSF

A 384-well ELISA microplate (Nunc MaxiSorb, Nunc) was coated by overnight incubation at 4°C with 0.5

µg/ml Syn-140 (sheep anti-α-syn polyclonal antibody) in 200 mM NaHCO3, pH 9.6 (50 µl/well). The plate

was then washed with PBST and incubated with 100 µl/well of blocking buffer for 2 hours at 37°C. After

washing, 50 µl of the CSF samples (thawed on ice and Tween-20 was added to a final concentration of

0.05%) was added to each well, and plates were incubated at 37°C for 2.5 hours. PS129 (mouse anti-pS129-

α-syn monoclonal antibody) diluted in blocking buffer (1:1K) were added to the appropriate wells, and

incubated at 37°C for 2 hours. Next, the plate was washed and incubated for 2 hours at 37°C with 50 µl/well

of species-appropriate secondary antibody (donkey anti-mouse IgG HRP (Jackson ImmunoResearch))

  81  

diluted in blocking buffer (1:20 K). After washing, the plate was incubated with 50 µl/well of an enhanced

chemiluminescent substrate (SuperSignal ELISA Femto, Pierce Biotechnology, Rockford, IL). The

chemiluminescence, expressed in relative light units, was immediately measured using VICTOR™ X3

multilabel plate reader (PerkinElmer). The standard curve for the ELISA assay was obtained using serial

dilutions of recombinant human p-S129-α-syn in artificial CSF (50 µl/well).

Development of ELISA for measuring o-α-syn in human CSF

A 384-well ELISA microplate (Nunc MaxiSorb, Nunc) was coated by overnight incubation at 4°C with

Syn-O2 (0.2 µg/ml) in 200 mM NaHCO3, pH 9.6 (50 µl/well). The plate was then washed with PBST and

incubated with 100 µl/well of blocking buffer for 2 hours at 37°C. After washing, 50 µl of the CSF samples

(thawed on ice and Tween-20 was added to a final concentration of 0.05%) was added to each well, and

plates were incubated at 37°C for 2.5 hours. FL-140 (rabbit polyclonal antibody, Santa Cruz Biotechnology,

Santa Cruz, CA, USA), diluted in blocking buffer at 1:1K, was added to the appropriate wells, and incubated

at 37°C for 2 hours. Next, the plate was washed and incubated for 2 hours at 37°C with 50 µl/well of goat

anti-rabbit IgG HRP (Jackson ImmunoResearch) diluted in blocking buffer (1:15 K). After washing, the

plate was incubated with 50 µl/well of an enhanced chemiluminescent substrate (SuperSignal ELISA

Femto, Pierce Biotechnology, Rockford, IL). The chemiluminescence, expressed in relative light units, was

immediately measured using VICTOR™ X3 multilabel plate reader (PerkinElmer). The standard curve for

the ELISA assay was obtained using serial dilutions of recombinant human o-α-syn in artificial CSF (50

µl/well).

AD CSF biomarkers

Concentrations of t-tau, p-tau and Aβ42 in CSF were determined using the sandwich ELISAs Innotest β-

Amyloid (1–42), Innotest h TAU-Ag™ and Innotest Phosphotau (181P) ™; (Fujirebio Diagnostics),

respectively, as described previously [33].

Study population

A description of this cross-sectional cohort has been previously published [34]. We included 46 patients

with PD that attended the outpatient clinic for movement disorders at the VU University Medical Center

(VUMC) between September 2008 and February 2011, and 48 self-declared age-matched HC who were

recruited via an advertisement in the periodical of the Dutch Parkinson Foundation. All patients with PD

fulfilled the United Kingdom Parkinson’s Disease Society Brain Bank clinical diagnostic criteria [35].

Patients were included only if they were able to understand the study aim and procedures. Mini-Mental

State Examination and/ or neuropsychological assessment in the patients did not indicate dementia. In the

controls, dementia was excluded using the Cambridge Cognitive Examination scale [36]. Patients and

controls underwent a standardized clinical assessment that included their medical history and a neurological

examination. Severity of parkinsonism and disease stage in the ‘on’ state were rated using the Unified

  82  

Parkinson’s Disease Rating Scale-Part-III (UPDRS-III) [37] and the modified Hoehn and Yahr (H&Y)

classification [38], respectively. All the samples were screened in a blinded fashion and were tested

randomly. All the results were confirmed using at least two independent experiments. A series of internal

controls were also run to check for run-to-run variations. All CSF samples with > 500 erythrocytes/µl were

excluded from analysis as traces of blood may influence CSF α-syn levels [39].

Data Analysis

Statistical analyses were performed using using GraphPad Prism (version 5.0) software and IBM SPSS

software 21. Continuous variables were described using medians and interquartile ranges. Categorical

variables were presented as count or percentages. Mann-Whitney U-test was used for comparisons between

PD and HC diagnostic groups. Correlations were calculated using Spearman’s Rho(r). Subjects with PD

and HC were matched for age (p = 0·002), but not for gender since no significant differences between CSF

biomarkers in males and females were found in PD or HC groups (p = 0·85). The accuracy of the diagnostic

value of the biomarkers was assessed based on the area under the curve (AUC) of the receiver operating

characteristic (ROC) curve [40]. Cut-off values were calculated using sensitivity and specificity values that

maximized Youden’s index (sensitivity + specificity − 1). To determine the diagnostic value of a

combination of multiple biomarkers, we performed a binary logistic regression analysis. Combinations of

biomarkers were tested to find the best fitting model in a forward stepwise mode, which was based on the

change in likelihood resulting from including the variable. Change in -2 log-likelihood statistics between

the models was tested using Chi-square (χ2) tests. Sensitivity and specificity values of the combinations of

biomarkers for PD diagnosis and progression values were also calculated.

Results Development and characterization of our anti-α-syn antibodies

Our mouse monoclonal antibody, Syn-O2, specifically recognizes early “soluble oligomers” and late

aggregates “amyloid fibrils” of α-syn (Figure 1A,B), and has a high binding affinity for o-α-syn (IC50 120

pM and KD 9.6 x 10-11 M) [24]. It exclusively bound to α-syn aggregates (amyloid fibrils and soluble

oligomers) without cross-reactivity with monomers or fibrils generated from other amyloid proteins

including β-syn, γ-syn, β-amyloid, tau, islet amyloid polypeptide (IAPP) or ABri (Figure 1A,B). In

immunohistochemical studies, Syn-O2 stained underappreciated small intra- and extracellular micro-

aggregates and very thin neurites in PD and DLB cases that were not observed with generic pan antibodies

(Syn-1 or KM51) that recognize linear epitope. Both classical Lewy neurites defined as elongated structures

within neuronal processes, and Lewy neurites in the form of beaded strings; swollen/bulging neurites and

long, thin threads were also stained by Syn-O2 (Figure 1C,D). The mAb,11D12, and a sheep polyclonal-

antibody, Syn-140, both recognized different forms/species of α-syn including monomeric-, oligomeric-,

protofibrillar-, nitrated- and phosphorylated-S129-α-syn, as well as full-length and N- or C-terminal

  83  

truncated forms of α-syn (Figure 1E-G). Finally, PS129, a mouse mAb was specific for p-S129-α-syn

(Figure 1H).

Development of new ELISA assays for measuring different species of α-syn

We developed, validated and optimized a new sandwich-type ELISA for measuring t-α-syn levels in human

We developed, validated and optimized a new sandwich-type ELISA for measuring t-α-syn levels in human

CSF. Assay conditions were first optimized using different concentrations of the capture antibody Syn-140

(sheep anti-α-syn polyclonal antibody). Based on its sensitivity and background signal, 0.1 µg/ml of the

capture antibody was selected for subsequent assays. The detection limit of the assay was as low as 50

pg/ml, which is 20-fold less than the concentration detected in human CSF (Figure 2A). The standard curve

for the ELISA assay was constructed using different concentrations of recombinant human α-syn in

solution. Serial dilutions of recombinant human β-syn and γ-syn were also included as negative controls

(Figure 2B). The inter-assay coefficient of variation (CV) examined over >10 plates was < 10% (Figure

2C). As illustrated in (Figure 2D), our assay permitted the direct quantification of native α-syn in biological

samples such as human CSF and Tg mouse-brain lysates. α-Syn was detected in Tg mice and human CSF,

whereas no signal was observed in WT or KO lysates (Figure 2D). Moreover, recovery rates were

determined by spiking recombinant α-syn into human CSF. Our assay consistently featured a mean recovery

rate > 90% (Figure 2E).

We followed a similar protocol for p-S129-α-syn-specific ELISA, where Syn-140 was used as the capture

antibody and PS129 was used as the reporter. To address the issue of specificity, serial dilutions of

recombinant human p-S129-α-syn and non-phosphorylated α-syn were tested, and signal was detected only

with the p-S129-α-syn (Figure 3A). Using similar criteria (sensitivity, reproducibility and background

signal), optimal conditions were adopted for the assay (Figure 3B). The limit of detection was as low as 20

pg/ml, and a standard curve with R2 of 0.999 was generated (Figure 3B), whereas, the inter-assay CV was

< 15% (Figure 3C). To explore the assay suitability for quantifying p-S129-α-syn in biological fluids,

multiple concentrations of recombinant human p-S129-α-syn were spiked into human CSF. The mean

recovery rates were between 95 and 97% (Figure 3D).

  84  

Figure 1. Characterization of anti-α-syn antibodies.

50 ng of monomers (M) and fibrils (F) of human α-syn, ABri, Islet amyloid polypeptide (IAPP), Abeta or tau (A), and β- or γ-syn (B) were spotted onto nitrocellulose membranes, and then probed with the indicated antibodies. (C) Representative photomicrographs illustrating histopathological features detected with Syn-O2 in substantia nigra pars compacta (SNpc), CA2 and deep layers of entorhinal cortex (dENT) or superficial layers of entorhinal cortex (sENT) of a PD patient (male, 84 years; Braak PD stage 6). 1) Lewy body and small bulgy neurites in SNpc; 2) neuron with punctated Syn-O2 immunoreactivity in SNpc. *Note the Syn-O2 immunoreactive beaded strings. 3) long thin threads and many small extracellular or synaptic Syn-O2 immunoreactivity; 4) a long thread in CA2 and many small neurites; 5) Lewy body pathology in deep layers of enthorinal cortex; 6) many small neurites and synaptic staining (Bar = 50 µm). (D) Photomicrographs showing immunoreactivity of Syn-O2, KM-51 and Syn-1 monoclonal antibodies in 10 µm thick paraffin-embedded sections of a control and PD patient. No staining was observed in the control with any of these mAbs. In PD patient, small and large LBs and LNs were observed with all three mAbs. With Syn-O2, many other long neurites and small extracellular or synaptic-like aggregates were stained. Bar = 50 µm. (E) Monomeric (m-α-syn), oligomeric (o-α-syn),

A B

D

C

1 2

4 5

3

6

FE G H

Figure 1

  85  

phosphorylated α-syn (p-Ser129-α-syn) and nitrated (n-α-syn) forms were used to test the cross-reactivity of our antibodies 11D12 and Syn-140· The generic commercial mAbs Syn-1 (BD Biosciences, 50ng/ml) for α-syn, and EP1536Y (Abcam) for p-Ser129-α-syn, were also included as controls. (F) 50 ng of recombinant α-, β- and γ-syn were loaded on SDS gels and transferred to nitrocellulose membranes for western blotting, and then probed with our antibodies or control antibodies as appropriate. Syn-1(BD Biosciences, 50ng/ml) for α-syn, anti-β-synuclein (8) (Santa Cruz Biotechnology, 1:2K) for β-syn and C-20 (Santa Cruz Biotechnology, 1:3K) for γ-syn. (G) 15 µg of human, mouse and rat brain lysates were used in western blotting to test the specificity of 11D12 and Syn-140· 50ng of recombinant human α-syn (rec.α-syn) was included as positive control. (H) 50 ng of recombinant human α-syn (H-α-syn) or mouse α-syn (M-α-syn), human phosphorylated α-syn (H-p-Ser129-α-syn) and mouse phosphorylated α-syn (M-p-S129-α-syn) were loaded on SDS gels and transferred to nitrocellulose membranes for western blotting, and the membranes were then probed with our mouse mAb (PS129) recognizes both human and mouse p-Ser129-α-syn or the generic commercial mAb Syn-1 as indicated.

  86  

Figure 2. Validation of ELISA specific for total α-syn.

(A) Histograms representing the optimization of the capture antibody (Syn-140) concentration. Based on the sensitivity and background signal (BG), 0.1 µg/ml of the capture antibody was selected for subsequent assays. (B) Standard curves displaying the specificities and sensitivities for α- (u), β- (n) and γ-syn (▲). (C) Data shown are representative of 3 independent experiments using serial dilutions of recombinant α-syn freshly prepared over 3 non-consecutive days. Measurements were taken in duplicate, and the results show the mean ± standard deviation for each point, and the inter-assay coefficient of variation (CV) was < 10%. (D) Antibody specificity determination using murine brain lysates and human

C

0

20000

40000

60000

80000

100000

120000

140000

5 2 1 0.5

RLU

/Sec

α-syn concentration (ng/ml)

Day 1 Day 2 Day 3 5 ng/ml CV : 2.1%2 ng/ml CV : 8.5%1 ng/ml CV : 9.5%0.5 ng/ml CV : 9.2%

B

y = -1711.2x2 + 31859x + 1533R² = 0.9993

0

20000

40000

60000

80000

100000

120000

140000

0 1 2 3 4 5 6

RLU

/Sec

protein concentration (ng/ml)

α-syn β-syn γ-syn

CSF 1 CSF 2 CSF 3

0.00

0.20

0.40

0.60

0.80

1.00

1.20

1.40

1.60

1.80

α-sy

n co

ncen

trat

ion

(ng/

ml)

unspiked + 0.5 + 0.2 + 0.05 unspiked + 0.5 + 0.2 + 0.05 unspiked + 0.5ng/ml

+ 0.2ng/ml

+ 0.05ng/ml

Average recovery rate94%

Average recovery rate92% Average recovery rate

94%

E

D

A

  87  

CSF; Tg, WT and KO brain lysates were diluted to 5µg/ml. Our assay permitted the direct quantification of native α-syn in biological samples such as human CSF and Tg mouse-brain lysates, whereas no signal was observed in WT or KO lysates. (E) Assessment of α-syn recovery rate in human CSF. The recovery rates were calculated from measured α-syn concentrations relative to spiked amounts, and the assay consistently featured a mean recovery rate > 90%.

As described above, we developed and optimized novel specific ELISA for measuring o-α-syn in human

CSF. Similarly, our oligomeric ELISA is based on a conventional sandwich ELISA system, where o-α-syn

is captured using our highly specific anti-o-α-syn mAb; Syn-O2. Multiple conditions were tested to assess

assay performance. Serial dilutions of recombinant human o-α-syn were run in duplicates. Based on optimal

sensitivity, specificity, reproducibility and background signal results, we selected a concentration of 0.2

µg/ml for coating (Figure 4B).

Our assay specificity towards o-α-syn was further validated as no signal was detected when serial dilutions

of α-syn monomers were included (Figure 4C). The intra-assay and inter-assay precision was < 10% (Figure

4D). To assess the specificity and sensitivity of our oligomeric-ELISA for o-α-syn in biological samples,

brain lysates and microdialysis fluid from young Tg mice that over-expressed human α-syn were used, and

WT and KO mice were also included. As anticipated, o-α-syn levels were significantly greater in Tg mice

compared with WT, whereas no signal was detected in KO mice (Figure 4E). In parallel, our assay

quantified the levels of o-α-syn in human CSF successfully (Figure 4E). Next, we spiked increasing

amounts of recombinant o-α-syn into human CSF samples. Interestingly, the average recovery rates were

> 95% (Figure 4F). We estimated that the lower limit of detection of recombinant o-α-syn using this ELISA

was as low as 10 pg/ml based on the initial concentration of the protein (5 ng/ml). Taken together, the

results show that our oligomeric-ELISA is specific, sensitive and reproducible, and it is thus suitable for

the quantification of o-α-syn in biological samples.

  88  

Figure 3. Validation of ELISA specific for p-S129-α-syn.

(A) Standard curves displaying the specificities and sensitivities for p-S129-α-syn (u) and non-phosphorylated α-syn (n). (B) Histograms representing the optimization of the capture antibody (Syn-140) concentration and the background signal (BG). (C) Examination of inter-assay variability. Measurements were taken in duplicate, and the results show the mean ± standard deviation for each point, and the inter-assay variability was found < 15%. (D) Assessment of α-syn recovery rate in human CSF. The recovery rates were calculated between 95 and 97% from measured α-syn concentrations relative to spiked amounts.

C

0

500000

1000000

1500000

2000000

2500000

5 2 1 0.5

RL

U/S

ec

p-S129-α-syn concentration (ng/ml)

Day 1 Day 2 Day 3 5 ng/ml CV : 10.7%2 ng/ml CV : 12.7%1 ng/ml CV : 14.2%0.5 ng/ml CV : 10.3%

D

CSF 1 CSF 2 CSF 3

0

100

200

300

400

500

600

700

800

p-S1

29-α

-syn

(pg/

ml)

unspiked + 0.5ng/ml

+ 0.2ng/ml

+ 0.05ng/ml

unspiked + 0.5ng/ml

+ 0.2ng/ml

+ 0.05ng/ml

unspiked + 0.5ng/ml

+ 0.2ng/ml

+ 0.05ng/ml

Average recovery rate95%

Average recovery rate97%Average recovery rate

95%

B

y = -32143x2 + 664616xR² = 0.9992

0

500000

1000000

1500000

2000000

2500000

3000000

0 1 2 3 4 5 6

RLU

/Sec

protein concentration (ng/ml)

p-S129-α-syn α-syn

A

  89  

Figure 4. Validation of ELISA specific for o-α-syn.

(A) Standard curves displaying the specificities and sensitivities for p-S129-α-syn (u) and non-phosphorylated α-syn (n). (B) Histograms representing the optimization of the capture antibody (Syn-140) concentration and the background signal (BG). (C) Examination of inter-assay variability. Measurements were taken in duplicate, and the results show the mean ± standard deviation for each point, and the inter-assay variability was found < 15%. (D) Assessment of α-syn recovery rate in human CSF. The recovery rates were calculated between 95 and 97% from measured α-syn concentrations relative to spiked amounts.

0

200000

400000

600000

800000

1000000

1200000

5 2 1 0.5 0.2 0.1 0.05 0.02 0.01

RLU/

Sec

o-α-syn (ng/ml)

Syn-O2 0.5 µg/ml Syn-O2 0.2 µg/ml Syn-O2 0.1 µg/ml

0

5000

10000

15000

20000

25000

30000

35000

40000

0.1 0.05 0.02 0.01

BA

E

0

100

200

300

400

500

600

700

800

900

1000

o-α-

syn

(pg/

ml)

KO WT Tg KO WT Tg CSF

Mouse brain lysates Mouse ECF Human

! !

! not- detectable

# 1 # 2 # 3 # 1 # 2 # 3 # 1 # 2 # 3 # 1 # 2 # 3 # 1 # 2 # 3 # 1 # 2 # 3 # 1 # 2 # 3

C

y = -2778.9x2 + 153091x + 1669.9R² = 0.9999

0

100000

200000

300000

400000

500000

600000

700000

800000

0 1 2 3 4 5 6

RLU/

Sec

protein concentration (ng/ml)

α-syn oligomers α-syn monomers

0

100000

200000

300000

400000

500000

600000

700000

800000

900000

1000000

5 2 1 0.5R

LU/S

eco-α-syn (ng/ml)

Day 1 Day 2 Day 3 5 ng/ml CV : 3.9%2 ng/ml CV : 4.9%1 ng/ml CV : 5.5%0.5 ng/ml CV : 7.4%

D

F

  90  

CSF biomarkers in diagnostic groups

We then measured the levels of different α-syn species in human CSF of an independent cross-sectional

cohort consisting of individuals diagnosed with PD (Table 1) and age-matched HC. Consistent with

previous reports [11, 14, 16, 22], CSF t-α-syn levels were significantly decreased in PD compared to HC

(p < 0.0001), whereas, o-α-syn levels were significantly higher in the PD group (p < 0.0001). CSF p-S129-

α-syn levels were elevated in PD compared to HC, with considerable overlap between the groups (p

<0.001). However, both p-S129-/t-α-syn and o-/t-α-syn ratios improved the discrimination of PD from HC

(p < 0.0001). CSF t-tau, p-tau and Aβ42 levels did not differ significantly between PD and HC groups

(Table 1).

Table 1. Demographics, clinical features and biomarkers. Median, interquartile ranges and p-values of

Mann-Whitney U test continuous variables, Count (percentage) for categorical ones.

  HC  (n=48)   PD  (n=46)   p-­‐values  Female  Male  

32  (66.6%)  16  (33.3%)  

18  (39%)  28  (61%)  

 0.85  

Age   63  (57-­‐67)   64  (57-­‐71)   0.7969  Disease  years   -­‐   4  (2-­‐8.3)   -­‐  H&Y  Stage   -­‐   2  (2-­‐2.5)   N.A  UPDRS-­‐  III   -­‐   23  (15.3-­‐27)   N.A  MMSE   29  (29-­‐30)   29  (28-­‐30)   0.0479  t-­‐α-­‐syn  (ng/ml)   1.6  (1.3-­‐2.2)   1.3  (1.2-­‐1.6)   <0.0001  p-­‐S129-­‐α-­‐syn  (pg/ml)   222  (180.5-­‐275)   261  (206.8-­‐296.3)   <0.001  o-­‐α-­‐syn  (pg/ml)   57  (36-­‐106.5)   116  (76-­‐170)   <0.0001  p-­‐S129/t-­‐α-­‐syn  %   13.7  (9.2-­‐18.5)   18.6  (15.7-­‐23.3)   <0.0001  o-­‐/t-­‐α-­‐syn  %   3.5  (2.3-­‐6.2)   8.9  (5.2-­‐12.2)   <0.0001  t-­‐tau  (pg/ml)   229  (162-­‐271.5)   190  (157.8-­‐274.3)   0.1710  p-­‐tau  (pg/ml)   41.5  (29.3-­‐50.3)   41.1  ±  16   0.5965  Aβ

42  (pg/ml)   995.5  (877.5-­‐1153)   966.5  (794-­‐1077)   0.6745  

  91  

Figure 5. Scatter plots showing the correlation between t-α-syn and t-tau, p-tau and between o-α-syn and Hoehn and Yahr score (H&Y) scores.

The correlation between CSF t-α-syn with t-tau (r =0·46, p < 0·01) and p-tau (r = 0·53, p < 0·001) in the PD group (A-B). The correlation between CSF o-α-syn with H&Y scores (r = -0·37, p < 0·05) in the PD group (C). The dotted line is the 95% prediction interval for the calculated regression line (solid line).

r= 0.46p<0.01

A

r= -0.37p<0.05

C

r= 0.53p<0.001

B

  92  

Figure 6. Use of receiver operating curves (ROC) for the levels of α-syn species in CSF.

ROC curves based on logistic regression analyses for the classification of PD patients versus HC based on various predictors and combination of predictors.

1-Specificty

Sens

itivi

ty

AUC: 0.70Sens: 65 %Spec: 74 %

t-α-synA

1-SpecifictySe

nsiti

vity

AUC: 0.77Sens: 89 %Spec: 52 %

o-α-synB

1-Specificty

Sens

itivi

ty

AUC: 0.67Sens: 65 %Spec: 54 %

p-S129-α-synC

1-Specificty

Sens

itivi

ty

o-/t-α-syn %

AUC: 0.82Sens: 68 %Spec: 85 %

D

1-Specificty

Sens

itivi

ty

p-S129-/t-α-syn %

AUC: 0.76Sens: 80 %Spec: 56 %

E

  93  

Cross sectional correlation analysis of α-syn species in CSF

A positive correlation with t-α-syn was found for both t-tau and p-tau (rs = 0.47, rs= 0.53 respectively, p <

0.00, Figure 5A, B) in the PD group. In contrast, an inverse significant correlation was observed between

p-S129-syn levels and p-tau (rs = -0.30, p =0.043) within the PD group. The levels of CSF o-α-syn did not

correlate with any of AD biomarkers (Table 2). While CSF t-α-syn did not correlate with H&Y, UPDRS-

III or disease duration, an inverse correlation with MMSE scores was observed (rs = -0.46, p < 0.001) within

the PD group (Table 3). Similarly, CSF p-S129-α-syn did not correlate with disease severity. On the other

hand, a significant inverse correlation was noted between CSF o-α-syn and H&Y scores (rs = -0.37, p =

0.015, Figure 5C) within PD group, but not with UPDRS, disease duration or MMSE scores. As anticipated,

t-tau and p-tau positively correlated with age (p = 0.003, p < 0.0001, respectively) and negatively correlated

with MMSE scores in the PD group (rs= -0.35, p = 0.01, rs = -0.38, p = 0.009 respectively), whereas, Aβ42

didn’t correlate significantly with any of the clinical parameters.

Table 2. Spearman correlations between CSF biomarkers in PD.

    t-­‐α-­‐syn   p-­‐α-­‐syn   o-­‐α-­‐syn   t-­‐Tau   p-­‐Tau   Aβ42  t-­‐α-­‐syn   1·∙00                      p-­‐S129-­‐α-­‐syn   0·∙002   1·∙00                  

o-­‐α-­‐syn   0·∙21   -­‐0·∙03   1·∙00              t-­‐Tau   0·∙46**   -­‐0·∙19   0·∙06   1·∙00        p-­‐Tau   0·∙53***   -­‐0·∙30*   0·∙14   0·∙86**   1·∙00      Aβ42   0·∙09   -­‐0·∙14   0·∙5284   0·∙31*   -­‐     1·∙00  

Logistic regression analysis

Binary logistic regression analysis revealed five CSF biomarkers as individual PD predictors: t-α-syn (p =

0.003), o-α-syn (p <0.0001), p-S129-α-syn (p < 0.0001), o-/t-α-syn ratio (p <0.0001) and p-S129/t-α-syn

ratio (p < 0.0001; Figure 6A-E). The combination of o-/t-α-syn, p-S129-α-syn and p-tau formed the best

fitting predictive model for discriminating PD patients from controls (Table 4). The sensitivity and

specificity of the combination of o-/t-α-syn ratio, p-S129-α-syn and p-tau was 79% and 67% (AUC= 0.86).

The performance of this predictive model was significantly better compared to o-/t-syn ratio, p-S129-syn

alone (See Table 4 for further details).

  94  

Table 3. Spearman correlations between CSF biomarkers, disease duration (years), UPDRS-III, Hoehn and Yahr stage and MMSE in PD.

Disease years UPDRS-III Hoehn and Yahr stage MMSE t-α-syn 0·06 0·12 0·18 -0·46** p-S129-α-syn 0·04 -0·16 -0·18 -0·05 o-α-syn -0·25 -0·19 -0·37* -0·01 p-S129/t-α-syn % -0·16 -0·22 0·002 0·39** o/t-α-syn % -0·25 -0·26 -0·31* 0·24

Table 4. Logistic regression analysis of CSF biomarkers between PD and HC

Predictor AUC Sensitivity Specificty p value t-α-syn 0·70 65 % 74 % 0·001 p-S129-α-syn 0·67 65 % 54 % < 0·0001 o-α-syn 0·77 89 % 52 % < 0·0001 p-S129/t-α-syn % 0·76 80% 56% < 0·0001 o-/t-α-syn % 0·82 68% 85% < 0·0001 o-/t-α-syn %, p-S129-α-syn & p-tau 0·86 79% 67% < 0·0001

Discussion The lack of reliable biomarkers is a major obstacle holding us back from accurately diagnosing PD or

monitoring its progression. Since the precise identification of toxic α-syn species is still unclear, we aimed

to generate antibodies capable to detect wide range of the different α-syn species including the pathogenic

species o-α-syn and p-S129-α-syn and then utilized these antibodies to develop total-, oligomeric- and p-

S129-ELISA systems capable to quantify specifically these species in different biological samples. Our

novel antibody, Syn-O2, can facilitate our ability to study o-α-syn and explore its toxic characteristics.

Moreover, Syn-O2 could provide insights into the possible strategies through which we can halt PD

progression or reverse o-α-syn toxicity. Applying different immunoassays, Syn-O2 was shown to be highly

selective for o-α-syn since it did not cross-react with aggregated forms of other amyloidogenic proteins

(tau, Abeta, IAPP and ABri) and it did not recognize the other synuclein proteins (β- or γ-syn). Moreover,

Syn-O2 clearly stained LBs and LNs only. Most interestingly, comparing Syn-O2 staining pattern with the

other commercial antibodies that are widely used in IHC (Syn-1, KM-51), Syn-O2 not only showed a lack

of cross-reactivity with synaptic α-syn, but also detected pathology not detectable by other pan antibodies

confirming its specificity toward α-syn pathology.

Both Syn-140 and 11D12 are specific for α-syn, and while Syn-140 recognizes α-syn from different species

(human, mouse and rat), 11D12 is specific only for human α-syn. Moreover, PS129 specifically recognizes

p-S129-α-syn and does not cross-react with non-phosphorylated α-syn. This diversity in the specifications/

characteristics of our antibodies imparts distinctive virtues to our ELISA assays. Therefore, our assays can

  95  

serve as powerful research tools to investigate the potential of α-syn species in different biological samples.

Furthermore, our new ELISA assays provide multiple improvements over other reported immunoassays.

First, our ELISA design using 384-well plates is perfectly compatible for accommodating multiple

replicates even with limited volume of the sample (50 µl/well). Second, the enhanced sensitivity shown by

our assays validates their suitability for the analysis of human CSF specimens. Our assays were shown to

be highly target specific based on several methods, including specificity validation of the employed

antibodies to their respective antigens, using brain and blood products from genetically modified mice (Tg,

WT, KO) and monitoring assay precisions. Our ELISAs are also robust since recovery rates from spike

experiments were > 90%.

Our group has previously described the development of an ELISA for quantifying o-α-syn in human CSF

using the same mouse mAb (211, mouse anti-α-syn antibody, Santa Cruz) for capture and the biotinylated

form of 211 for detection [14, 41]. However, heterophilic antibodies may interfere in this assay format

when using the same antibody for capturing and detection, leading to false-positive signals [42]. Our new

oligomeric-ELISA has addressed this shortcoming by using our novel mouse conformation-specific mAb

(Syn-O2), which selectively captures o-α-syn, and a rabbit polyclonal antibody (FL-140) for detection, thus

permitting the precise quantification of o-α-syn in biological samples. Furthermore, our new oligomeric-

ELISA system showed great sensitivity and specificity for measuring α-syn oligomers in biological samples

in comparison to our previous oligomeric-ELISA system.

Analyses of the cross sectional cohort revealed that while concentrations of t-α-syn decreased, o-α-syn

concentrations significantly increased in PD compared to HC, consistent with other studies [11, 12, 16, 34,

43, 44]. The reduction in CSF t-α-syn is likely due to α-syn oligomerisation and sequestration in LBs [11,

45], and we speculate that the elevated levels of o-α-syn results from a clearance failure of the aggregated

forms of α-syn. It’s worth mentioning, that in a recent study by Compta et al [44], the levels of o-α-syn

were determined using our previously published oligomeric-ELISA [14], where o-α-syn levels were found

to be elevated in PD patients consistent with our findings in this study. However, we attribute the differences

in the levels between both studies to the use of different quantification methods, pre-analytical confounding

factors (types of tubes, spinning conditions, storage conditions, etc.) and differences in disease severity

between the patients included in the studies. Furthermore, as emphasized above, we found our new

oligomeric-ELISA to be more reliable compared to the old generation oligomeric-ELISA in terms of

sensitivity, specificity and robustness.

PD patients with dementia were not enrolled in our cohort, as reflected in the overall high MMSE scores

registered. These high MMSE scores could explain the weakness of the correlation between MMSE scores

and t-α-syn levels and the lack of a correlation with Aβ42 levels. Interestingly, CSF o-α-syn levels showed

modest negative correlation with disease motor severity (H&Y grade). This inverse correlation, although

  96  

weak, suggests that o-α-syn increases at the early stage of the disease prior to the manifestation of the motor

symptoms. Interestingly, similar finding has been recently reported in sporadic PD patients and PD cases

with leucine-rich repeat kinase 2 mutations [23].

An increase in p-S129-α-syn levels was also observed in the PD group compared to HC. Although,

approximately 90% of accumulated α-syn in LBs consists of p-S129-α-syn [46], it is not known whether

phosphorylation of α-syn promotes or prevents the formation of toxic α-syn aggregates. To date there is

only one reported study, in which there was a weak positive correlation between CSF p-S129-α-syn/t-α-

syn ratio and disease severity [15], however we did not observe such relationship in our current study. These

discrepancies are likely due to several factors, including differences in the assay platform (Luminex versus

sandwich-ELISA), collection and handling of the CSF samples and/or heterogeneity of the patients included

in the studies.

Nevertheless, including both o-/t-α-syn and p-S129-/t-α-syn ratios improved the ability to discriminate

between PD and HC, this was further enhanced by including p-tau, emphasizing the usefulness of

combining several CSF biomarkers for diagnostic purposes.

Synucleinopathies and tauopathies show significant clinical overlap, making the early diagnosis of PD more

challenging. This overlap necessitates a combination of measurements of CSF α-syn species with key AD

biomarkers to improve diagnostic accuracy. Moreover, several in vitro and in vivo studies investigated

interactions among α-syn and tau proteins, showing that these proteins promote each other’s aggregation,

leading to neuronal degeneration and worsening cognitive impairment [47]. The positive correlation

between CSF t-α-syn and t-tau and p-tau in the PD group we observed, confirms and extends reports by

others showing a positive correlation between CSF t-α-syn and tau levels in PD, AD and controls [48] .

Conclusions

In summary, using our novel mAbs, we have developed sensitive and specific ELISAs to measure t-, o- and

p-S129-α-syn species in human CSF. Combining measurements of different α-syn species in CSF, we

observed marked differential CSF patterns between PD and controls. Our results validate the usefulness of

combining multiple CSF biomarkers in improving PD diagnostic accuracy and prognosis. Although the

potential use of our assays was confirmed in an independent cohort, we still need to validate the utilization

of our assays in large-scale, prospective, longitudinal and well-controlled studies such as the ongoing

Parkinson’s Progression Markers Initiative, and by inclusion of patients with other neurological illnesses.

  97  

References [1] J. Jankovic, "Parkinson's disease: clinical features and diagnosis," (in eng), J Neurol Neurosurg Psychiatry, vol. 79, no. 4, pp. 368-76, Apr 2008. [2] A. DelleDonne et al., "Incidental Lewy body disease and preclinical Parkinson disease," (in eng), Arch Neurol, vol. 65, no. 8, pp. 1074-80, Aug 2008. [3] J. M. Milber et al., "Lewy pathology is not the first sign of degeneration in vulnerable neurons in Parkinson disease," (in eng), Neurology, vol. 79, no. 24, pp. 2307-14, Dec 11 2012. [4] E. Tolosa, C. Gaig, J. Santamaría, and Y. Compta, "Diagnosis and the premotor phase of Parkinson disease," (in eng), Neurology, vol. 72, no. 7 Suppl, pp. S12-20, Feb 2009. [5] A. A. Dijkstra et al., "Stage-dependent nigral neuronal loss in incidental Lewy body and Parkinson's disease," (in eng), Mov Disord, vol. 29, no. 10, pp. 1244-51, Sep 2014. [6] A. Anoop, P. K. Singh, R. S. Jacob, and S. K. Maji, "CSF Biomarkers for Alzheimer's Disease Diagnosis," (in eng), Int J Alzheimers Dis, vol. 2010, 2010. [7] M. G. Spillantini, M. L. Schmidt, V. M. Lee, J. Q. Trojanowski, R. Jakes, and M. Goedert, "Alpha-synuclein in Lewy bodies," (in eng), Nature, vol. 388, no. 6645, pp. 839-40, Aug 28 1997. [8] K. E. Paleologou, G. B. Irvine, and O. M. El-Agnaf, "Alpha-synuclein aggregation in neurodegenerative diseases and its inhibition as a potential therapeutic strategy," (in eng), Biochem Soc Trans, vol. 33, no. Pt 5, pp. 1106-10, Nov 2005. [9] O. M. el-Agnaf, G. B. Irvine, and D. J. Guthrie, "Conformations of beta-amyloid in solution," (in eng), J Neurochem, vol. 68, no. 1, pp. 437-9, Jan 1997. [10] O. M. El-Agnaf, D. M. Walsh, and D. Allsop, "Soluble oligomers for the diagnosis of neurodegenerative diseases," (in eng), Lancet Neurol, vol. 2, no. 8, pp. 461-2, Aug 2003. [11] T. Tokuda et al., "Decreased alpha-synuclein in cerebrospinal fluid of aged individuals and subjects with Parkinson's disease," (in eng), Biochem Biophys Res Commun, vol. 349, no. 1, pp. 162-6, Oct 2006. [12] B. Mollenhauer, J. J. Locascio, W. Schulz-Schaeffer, F. Sixel-Döring, C. Trenkwalder, and M. G. Schlossmacher, "α-Synuclein and tau concentrations in cerebrospinal fluid of patients presenting with parkinsonism: a cohort study," (in eng), Lancet Neurol, vol. 10, no. 3, pp. 230-40, Mar 2011. [13] L. Parnetti et al., "Cerebrospinal fluid Tau/α-synuclein ratio in Parkinson's disease and degenerative dementias," (in eng), Mov Disord, vol. 26, no. 8, pp. 1428-35, Jul 2011. [14] T. Tokuda et al., "Detection of elevated levels of α-synuclein oligomers in CSF from patients with Parkinson disease," (in eng), Neurology, vol. 75, no. 20, pp. 1766-72, Nov 2010. [15] Y. Wang et al., "Phosphorylated α-synuclein in Parkinson's disease," (in eng), Sci Transl Med, vol. 4, no. 121, p. 121ra20, Feb 2012. [16] L. Parnetti et al., "Differential role of CSF alpha-synuclein species, tau, and Aβ42 in Parkinson's Disease," (in eng), Front Aging Neurosci, vol. 6, p. 53, 2014. [17] K. D. van Dijk, M. Bidinosti, A. Weiss, P. Raijmakers, H. W. Berendse, and W. D. van

  98  

de Berg, "Reduced alpha-synuclein levels in cerebrospinal fluid in Parkinson's disease are unrelated to clinical and imaging measures of disease severity," (in eng), Eur J Neurol, vol. 21, no. 3, pp. 388-94, Mar 2014. [18] E. Rockenstein et al., "Accumulation of oligomer-prone α-synuclein exacerbates synaptic and neuronal degeneration in vivo," (in eng), Brain, vol. 137, no. Pt 5, pp. 1496-513, May 2014. [19] K. E. Paleologou et al., "Detection of elevated levels of soluble alpha-synuclein oligomers in post-mortem brain extracts from patients with dementia with Lewy bodies," (in eng), Brain, vol. 132, no. Pt 4, pp. 1093-101, Apr 2009. [20] R. Sharon, I. Bar-Joseph, M. P. Frosch, D. M. Walsh, J. A. Hamilton, and D. J. Selkoe, "The formation of highly soluble oligomers of alpha-synuclein is regulated by fatty acids and enhanced in Parkinson's disease," (in eng), Neuron, vol. 37, no. 4, pp. 583-95, Feb 2003. [21] O. Hansson et al., "Levels of cerebrospinal fluid α-synuclein oligomers are increased in Parkinson's disease with dementia and dementia with Lewy bodies compared to Alzheimer's disease," (in eng), Alzheimers Res Ther, vol. 6, no. 3, p. 25, 2014. [22] L. Parnetti et al., "Cerebrospinal fluid lysosomal enzymes and alpha-synuclein in Parkinson's disease," (in eng), Mov Disord, vol. 29, no. 8, pp. 1019-27, Jul 2014. [23] J. O. Aasly et al., "Elevated levels of cerebrospinal fluid α-synuclein oligomers in healthy asymptomatic LRRK2 mutation carriers," (in eng), Front Aging Neurosci, vol. 6, p. 248, 2014. [24] N. N. Vaikath et al., "Generation and characterization of novel conformation-specific monoclonal antibodies for α-synuclein pathology," (in eng), Neurobiol Dis, vol. 79, pp. 81-99, Jul 2015. [25] M. T. Ardah et al., "Structure activity relationship of phenolic acid inhibitors of α-synuclein fibril formation and toxicity," (in eng), Front Aging Neurosci, vol. 6, p. 197, 2014. [26] O. M. El-Agnaf et al., "Effect of the disulfide bridge and the C-terminal extension on the oligomerization of the amyloid peptide ABri implicated in familial British dementia," (in eng), Biochemistry, vol. 40, no. 12, pp. 3449-57, Mar 2001. [27] A. M. Bodles, O. M. El-Agnaf, B. Greer, D. J. Guthrie, and G. B. Irvine, "Inhibition of fibril formation and toxicity of a fragment of alpha-synuclein by an N-methylated peptide analogue," (in eng), Neurosci Lett, vol. 359, no. 1-2, pp. 89-93, Apr 2004. [28] S. Poole, A. F. Bristow, S. Selkirk, and B. Rafferty, "Development and application of radioimmunoassays for interleukin-1 alpha and interleukin-1 beta," (in eng), J Immunol Methods, vol. 116, no. 2, pp. 259-64, Jan 1989. [29] Y. S. Taktak et al., "Assay of pyrogens by interleukin-6 release from monocytic cell lines," (in eng), J Pharm Pharmacol, vol. 43, no. 8, pp. 578-82, Aug 1991. [30] J. H. Lu et al., "Baicalein inhibits formation of α-synuclein oligomers within living cells and prevents Aβ peptide fibrillation and oligomerisation," (in eng), Chembiochem, vol. 12, no. 4, pp. 615-24, Mar 2011. [31] A. Masad et al., "Copper-mediated formation of hydrogen peroxide from the amylin peptide: a novel mechanism for degeneration of islet cells in type-2 diabetes mellitus?," (in eng), FEBS Lett, vol. 581, no. 18, pp. 3489-93, Jul 2007.

  99  

[32] M. T. Ardah et al., "Ginsenoside Rb1 inhibits fibrillation and toxicity of alpha-synuclein and disaggregates preformed fibrils," (in eng), Neurobiol Dis, vol. 74, pp. 89-101, Feb 2015. [33] C. Mulder et al., "Amyloid-beta(1-42), total tau, and phosphorylated tau as cerebrospinal fluid biomarkers for the diagnosis of Alzheimer disease," (in eng), Clin Chem, vol. 56, no. 2, pp. 248-53, Feb 2010. [34] K. D. van Dijk, M. Bidinosti, A. Weiss, P. Raijmakers, H. W. Berendse, and W. D. van de Berg, "Reduced α-synuclein levels in cerebrospinal fluid in Parkinson's disease are unrelated to clinical and imaging measures of disease severity," (in eng), Eur J Neurol, vol. 21, no. 3, pp. 388-94, Mar 2014. [35] A. J. Hughes, S. E. Daniel, L. Kilford, and A. J. Lees, "Accuracy of clinical diagnosis of idiopathic Parkinson's disease: a clinico-pathological study of 100 cases," (in eng), J Neurol Neurosurg Psychiatry, vol. 55, no. 3, pp. 181-4, Mar 1992. [36] M. Roth et al., "CAMDEX. A standardised instrument for the diagnosis of mental disorder in the elderly with special reference to the early detection of dementia," (in eng), Br J Psychiatry, vol. 149, pp. 698-709, Dec 1986. [37] Fahn, "Unified Parkinson’s disease rating scale. In: Fahn S, Marsden C, Calne D, eds. Recent Development in Parkinson’s Disease. ," vol. 2, R. Elton, Ed., ed. MacMillan Health Care Information, 1987. [38] J. Jankovic et al., "Variable expression of Parkinson's disease: a base-line analysis of the DATATOP cohort. The Parkinson Study Group," (in eng), Neurology, vol. 40, no. 10, pp. 1529-34, Oct 1990. [39] R. Barbour et al., "Red blood cells are the major source of alpha-synuclein in blood," (in eng), Neurodegener Dis, vol. 5, no. 2, pp. 55-9, 2008. [40] K. Hajian-Tilaki, "Receiver Operating Characteristic (ROC) Curve Analysis for Medical Diagnostic Test Evaluation," (in ENG), Caspian J Intern Med, vol. 4, no. 2, pp. 627-635, 2013. [41] O. M. El-Agnaf et al., "Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson's disease," (in eng), FASEB J, vol. 20, no. 3, pp. 419-25, Mar 2006. [42] D. Sehlin et al., "Interference from heterophilic antibodies in amyloid-β oligomer ELISAs," (in eng), J Alzheimers Dis, vol. 21, no. 4, pp. 1295-301, 2010. [43] M. J. Park, S. M. Cheon, H. R. Bae, S. H. Kim, and J. W. Kim, "Elevated levels of α-synuclein oligomer in the cerebrospinal fluid of drug-naïve patients with Parkinson's disease," (in eng), J Clin Neurol, vol. 7, no. 4, pp. 215-22, Dec 2011. [44] Y. Compta et al., "Correlates of cerebrospinal fluid levels of oligomeric- and total-α-synuclein in premotor, motor and dementia stages of Parkinson's disease," (in eng), J Neurol, vol. 262, no. 2, pp. 294-306, Feb 2015. [45] T. Stewart et al., "Cerebrospinal Fluid α-Synuclein Predicts Cognitive Decline in Parkinson Disease Progression in the DATATOP Cohort," (in eng), Am J Pathol, vol. 184, no. 4, pp. 966-75, Apr 2014. [46] J. P. Anderson et al., "Phosphorylation of Ser-129 is the dominant pathological

  100  

modification of alpha-synuclein in familial and sporadic Lewy body disease," (in eng), J Biol Chem, vol. 281, no. 40, pp. 29739-52, Oct 2006. [47] S. Moussaud, D. R. Jones, E. L. Moussaud-Lamodière, M. Delenclos, O. A. Ross, and P. J. McLean, "Alpha-synuclein and tau: teammates in neurodegeneration?," (in eng), Mol Neurodegener, vol. 9, p. 43, 2014. [48] S. Slaets et al., "Increased CSF α-synuclein levels in Alzheimer's disease: correlation with tau levels," (in eng), Alzheimers Dement, vol. 10, no. 5 Suppl, pp. S290-8, Oct 2014.

  101    

!!

CHAPTER 4

Longitudinal Changes in CSF Alpha-Synuclein Species Reflect Parkinson’s Disease Progression

  102  

CHAPTER 4 | LONGITUDINAL CHANGES IN CSF ALPHA-SYNUCLEIN SPECIES REFLECT PARKINSON’S DISEASE PROGRESSION Nour K. Majbour, MSc1, 2, Nishant N. Vaikath, MSc1, 3, Paolo Eusebi, PhD4, Davide Chiasserini, PhD4, Mustafa Ardah, PhD5, Shiji Varghese, MSc5, Takahiko Tokuda, M.D, PhD5, Peggy Auinger, MSc6, Paolo Calabresi, M.D, PhD4, 7, Lucilla Parnetti, M.D, PhD4, and Omar M.A El-Agnaf, PhD1,9 1Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 5825, Doha, Qatar. 2Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU University Medical Centre, Amsterdam, The Netherlands. 3Neural Plasticity and Repair Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, BMC A10, Lund University, Lund, Sweden 4Dipartimento di Medicina, Sezione di Neurologia, Università degli Studi di Perugia, Perugia, Italy 5Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates. 6Department of Neurology, Research Institute for Geriatrics, Kyoto Prefectural University of Medicine, Kyoto, 602-0841, Japan 7Department of Neurology, Center for Human Experimental Therapeutics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA 8IRCCS - S. Lucia Foundation, Rome, Italy 9Life Sciences Division, College of Science and Engineering, Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, PO Box 5825, Doha, Qatar. [email protected]. Mov Disord. 2016 Oct;31(10):1535-1542. doi: 10.1002/mds.26754.

     

  103  

ABSTRACT

Background: Parkinson’s disease (PD) diagnosis is mainly based on clinical criteria with high risk of

misdiagnosis. Identification of reliable biomarkers for disease diagnosis and progression has a key role for

developing disease-modifying therapies. Here we investigate the longitudinal changes of CSF α-synuclein

(α-syn) species in early PD patients, and explore the potential use of these species as surrogate biomarkers

for PD progression.

Methods: We used our newly developed ELISA systems for measuring different forms of α-syn, such as

oligomeric-α-syn (o-α-syn), phosphorylated-α-syn at serine 129 (p-S129-α-syn) or total-α-syn (t-α-syn) in

CSF from longitudinal DATATOP cohort (n = 121). CSF Alzheimer’s disease biomarkers (total-tau,

phosphorylated-tau, Aβ-40 and Aβ-42) were also measured for this cohort.

Results: Interestingly, t- and o-α-syn levels significantly increased over the two-year DATATOP follow-

up period, whereas p-S129-α-syn levels showed a longitudinal decrease. We have also noted a significant

association between o-/t-α-syn ratio and UPDRS motor change (r=-0.22, p<0.05) in all patients, which was

found to be stronger in postural instability and gait difficulty dominant PD group (r=-0.41, p<0.01), while

it was not significant in the tremor dominant subtype (r=-0.05, p=0.782). A strong positive correlation

between the changes in CSF t-α-syn and o-α-syn over the two-year DATATOP study was also noted (r =

0.8).

Conclusion: Together, our data show that CSF α-syn species have a dynamic pattern along the course of

the disease, suggesting their possible role as progression biomarkers for PD. Moreover, our findings seem

to suggest that the PD clinical phenotypes could be characterized by different CSF patterns.

Keywords: Parkinson’s disease; alpha-synuclein; oligomers; biomarkers; DATATOP.

INTRODUCTION

Parkinson’s disease (PD) is characterized by a long preclinical phase between the onset of the

neurodegeneration and the first appearance of motor manifestations [1]. Reliable biomarkers are urgently

needed to aid earlier disease diagnosis and prognosis, especially during the premotor stage. Aggregated and

hyper-phosphorylated forms of alpha-synuclein (α-syn) are the major components of Lewy bodies (LBs)

the hallmark pathology of PD [2]. Although abnormal aggregation of α-syn plays a critical role in PD

pathogenesis, the exact pathogenic α-syn species remain undefined[3]. Several studies have demonstrated

that amyloid oligomers, but not mature amyloid fibrils, are the neurotoxic species [4, 5]. Therefore, several

studies have explored CSF α-syn as a putative biomarker for PD. Trends for lower levels of CSF total-α-

syn (t-α-syn), higher α-syn oligomers (o-α-syn) levels and/or a marginally higher p-S129-α-syn/t-α-syn

ratio in PD have been reported, with a large overlap between PD patients and controls [6-12].

To fully explore the potential use of CSF α-syn species as biomarkers for the diagnosis and prognosis of

PD, robust methods are needed to accurately quantify specific forms of α-syn in biological fluids, and large-

  104  

scale longitudinal studies are also needed. To address this need, recently we developed robust and specific

ELISAs to assess different species of α-syn (t-, o- or p-S129-α-syn) in biological fluids[12, 13]. In this

study we used our new ELISAs to explore the potential use of CSF α-syn species as progression biomarkers

in 121 early PD patients from the longitudinal DATATOP (deprenyl and tocopherol antioxidative therapy

for Parkinsonism) study, who underwent repeated CSF sampling over a two-year period.

MATERIALS AND METHODS

The standard for the oligomeric ELISA

Expression and purification of recombinant human α-syn was performed as previously described [12, 14].

α-Syn oligomers were prepared by incubating fresh α-syn (25 µM) with Ginsenoside Rb1 (100 µM) in

PBS at 37°C for 5 days with continuous shaking at 800 rpm in a Thermomixer (Eppendorf), then α-syn

oligomers were characterized using electron microscopy as previously described [15]. Briefly, size

exclusion chromatography was carried out using an AKTA FPLC system (GE Healthcare, Sweden) and a

Superdex 200 column at 4 °C, in order to separate soluble α-syn oligomers from monomers. This way, we

guaranteed that only soluble α-syn oligomers were separated and then confirmed by electron microscopy

[15].

ELISA assays for measuring CSF α-syn species

CSF t-, o- and p-S129-α-syn levels were measured using our newly developed ELISA assays [12]. Briefly,

to capture t-α-syn or p-S129-α-syn, a 384-well ELISA microplate was coated by overnight incubation at

4°C with 0.1 µg/ml Syn-140 (sheep anti-α-syn polyclonal antibody) in 200 mM NaHCO3, pH 9.6 (50

µl/well), and Syn-O2 (0.2 µg/ml), was used to capture α-syn oligomers. The plates were then washed with

phosphate-buffered saline containing 0.05% Tween-20 (PBST) and incubated with 100 µl/well of blocking

buffer (PBST containing 2.5% gelatin) for 2 hours at 37°C. After washing, 50 µl of the CSF samples

(thawed on ice and Tween-20 was added to a final concentration of 0.05%) was added to each well, and

plates were incubated at 37°C for 2.5 hours. 11D12 (mouse anti-α-syn monoclonal antibody), PS129

(mouse anti-pS129-α-syn monoclonal antibody) and FL-140 (rabbit polyclonal antibody, Santa Cruz

Biotechnology, Santa Cruz, CA, USA) for measuring t-α-syn, pS129-α-syn or o-α-syn, respectively, diluted

in blocking buffer at 1:5 K, 1:1K and 1:1K, respectively, were added to the appropriate wells , and

incubated at 37°C for 2 hours. Next, the plates were washed and incubated for 2 hours at 37°C with 50

µl/well of species-appropriate secondary antibodies (donkey anti-mouse IgG HRP or goat anti-rabbit IgG

HRP (Jackson ImmunoResearch)) diluted in blocking buffer (1:20 K). After washing, the plates were

incubated with 50 µl/well of an enhanced chemiluminescent substrate (SuperSignal ELISA Femto, Pierce

Biotechnology, Rockford, IL). The chemiluminescence, expressed in relative light units, was immediately

measured using VICTOR™ X3 multilabel plate reader (PerkinElmer). The standard curve for the ELISA

  105  

assays was obtained using serial dilutions of recombinant human α-syn, p-S129-α-syn or o-α-syn in

artificial CSF (50 µl/well). Samples were measured in a blinded fashion and tested randomly. The baseline

and follow-up samples from the same individual were run in one plate to avoid plate-to-plate variations,

and results were confirmed with at least two independent experiments. A series of internal controls was

also run to check for run-to-run variations.

AD CSF biomarkers

AD biomarkers were measured with the INNO-BIA ALZBIO3 kit (Fujirebio Diagnostics), as previously

reported [16].

Study population

A description of the DATATOP cohort has been previously published [17]. DATATOP is a multicenter,

placebo-controlled clinical trial in patients with early PD. Eligible patients with early stages of untreated

PD were enrolled in DATATOP and randomized to (1) active deprenyl, (2) active tocopherol, (3) active

deprenyl and tocopherol, or (4) placebo treatments. Subjects presented at baseline with minimal disability,

which did not necessitate the need for symptomatic anti-PD treatment. Subjects were followed for two years

to an “endpoint” defined as the development of symptoms of sufficient severity to require dopamine

replacement therapy. Both CSF and the clinical data investigated in this study were collected for the patients

at the baseline and follow-up time points, prior to dopamine replacement therapy.

The criteria behind the clinical diagnosis were derived by the DATATOP Steering Committee members

who are experienced PD investigators. In selecting the CSF samples for the α-syn oligomers study, The

Parkinson’s’ study Group (PSG) committee excluded subjects that likely did not have PD. More

specifically, based on the investigators report, only subjects that had at least 60% confidence that PD was

the likely diagnosis are part of the study. This has been a typical cutoff in other studies to select definite

and acceptable PD. These subjects were divided into 90-100% confidence versus at least 60% confidence

at each of the visits. However, a confirmation of 100% likely PD was not feasible, since only a small number

(9 of the 800 subjects) of subjects where identified by autopsy whether the subject definitely had PD or not.

However, none of the 147 subjects in our study reported autopsy results. Thus, we have selected only 121

subjects whom did have definite PD (90-100% confidence). Investigators could only identify ranges of

confidence and 90-100% were grouped together for this study.

Clinical Assessment’s

Patients were classified into three subtypes following the approach of Jankovic and colleagues [18].

Depending on the dominant motor features, PD subtype was defined as (i) tremor dominant; (ii) postural

instability and gait difficulty dominant (PIGD); or (iii) intermediate. Subtype classification was based on

the ratio of the average of UPDRS tremor items (16, 20 and 21) divided by the average of ‘postural

  106  

instability and gait difficulty dominant’ items (13–15, 29 and 30). This ratio indicates PD subtypes as

follows: ≤ 1.0 = PIGD PD, ≥1.5 = tremor dominant >1.0 and <1.5 = intermediate.

In our study, this has resulted in 45 tremor dominant, 54 postural instability and gait difficulty dominant

and 21 intermediate cases (Table 1).

Furthermore, UPDRS scores were divided into sub-scores for tremor, rigidity, bradykinesia, and axial

impairment[19]: the sum of UPDRS items 20 and 21 for the tremor score, item 22 for the rigidity score, the

sum of items 24, 25, 26, and 31 for the bradykinesia score, the sum of items 27, 28, 29, and 30 for the axial

impairment score (arising from chair, posture, gait and postural stability), and the sum of items 18 and 19

for the bulbar score (speech and facial expression).

Data Analysis

Statistical analyses were performed using IBM SPSS software 21 and R software version 3.1. Continuous

variables were described using medians and interquartile ranges. Categorical variables were presented as

count or percentages. Differences between baseline and follow-up were assessed using Mann-Whitney U-

test for paired comparisons. Correlations were calculated using Spearman’s Rho(r). Multivariate linear

models were performed for testing the confounding effects of follow-up time. We have also tested the effect

of age and gender on the measurements of CSF biomarkers by means of two-way ANOVA.

Table 1. Demographics and clinical features at baseline. Median and interquartile ranges for

continuous variables, count (percentage) for categorical ones.

Variable Age (yrs) 60.0 (34.0-79.0) Disease duration (yrs) 0.6 (0.1-4.5) Follow-up (months) 31 (20-37) H&Y 1.5 (1.0-2.0) UPDRS-motor 12.2 (0.0-35.0) ADL 5.5 (4-8) Treatment Placebo 26 (21.5) Deprenyl 39 (32.2) Tocopherol 26 (21.5) Deprenyl + Tocopherol 30 (24.8) Motor Subtypes Tremor dominant 45 Postural instability gait dominant 54 Intermediate 21

  107  

RESULTS Recently, we developed three robust, sensitive and reproducible sandwich-type ELISA systems that were

specific for t-, p-S129- or o-α-syn, and suitable for quantifying α-syn species/forms in human CSF

samples[12]. The total-ELISA system permitted quantification of most native forms of α-syn present in

biological samples such as human CSF and human- and Tg mouse-brain lysates, and had a detection limit

of 50 pg/ml, which is 20-fold less than the concentration detected in human CSF. The inter-assay coefficient

of variation (CV) examined over >10 plates was < 10%. Moreover, recovery rates were determined by

spiking recombinant α-syn into human CSF. Our assay consistently featured a mean recovery rate > 90%.

For the p-S129-α-syn-specific ELISA, the limit of detection was as low as 20 pg/ml, and a standard curve

with R2 of 0.999 was generated, whereas, the inter-assay CV was < 15% .The mean recovery rates were

between 95 and 97%, while the new oligomeric-ELISA for measuring o-α-syn had a detection limit of 10

pg/ml [20] . Our assay specificity towards o-α-syn was further validated as no signal was detected when

serial dilutions of α-syn monomers were included. The intra-assay and inter-assay precision was < 10%.

Spiking increasing amounts of recombinant o-α-syn into human CSF samples, the average recovery rates

were > 95%. Thus in the current study we employed our new ELISA protocols to quantify t-, p-S129- and

o-α-syn in CSF samples from longitudinal DATATOP cohort.

Assessment of longitudinal changes of α-syn species in CSF

CSF α-syn, tau and Aβ species were measured in CSF samples taken from the large cohort of 121

participants in the DATATOP study who were clinically diagnosed with early PD at the last evaluation

(probability between 90% - 100%). We tested the change in CSF t-, o-, p-S129-α-syn, t-tau, p-tau, Aβ40

and Aβ42 over the two-year DATATOP follow-up period. Examining the effect of medication between the

different treatment groups, no significant effect was noted on the clinical outcomes. However, deprenyl

was found to significantly, although weakly, slow ADL loss. Further investigating the association between

the different treatments and the changes in CSF biomarkers, no significant effect was noted, eliminating

the assumption that medications are potential confounders. Testing for age and gender effect, a significant

increase of p-S129-α-syn was noted in males versus females, as well as a significant increase of t-tau and

p-tau with age. Thus, we have adjusted for age and gender when investigating the correlations between the

levels of the biomarkers and the clinical parameters. However, when testing the effect of age and gender

on the changes of CSF biomarkers by means of two-way ANOVA, no significant association was observed,

thus age and gender were not considered as potential confounders in investigating the changes of CSF

biomarkers and the change in the clinical outcome. Both CSF t-α-syn and o-α-syn levels and the o-/t-α-syn

ratio were significantly higher at follow-up compared to baseline (p =0.003, p <0.0001, and p <0.0001,

  108  

respectively). In contrast, p-S129-α-syn levels were significantly lower at follow-up compared to baseline

(p=0.001). Biomarkers values at baseline and follow-up are all presented in Table 2.

Table 2. Biomarkers values at baseline and follow-up. Median and interquartile ranges Pairwise

Mann-Whitney test.

Biomarker Baseline Follow-up p-value t-α-syn (ng/ml) 2.6 (0.5-16.6) 3.3 (0.6-23.9) 0.003 p-S129-α-syn (pg/ml) 220 (51.7-642.1) 180 (12.0-542.8) 0.001 o-α-syn (pg/ml) 114 (0.0-652.6) 181 (37.9-790.0) <0.001 o-/t-α-syn 43.4 (11.3-543. 4) 48.6 (7.6-124.8) <0.001 p-S129-/t-α-syn 86.5 (6.2-540.0) 47.6 (1.1-481.9) <0.001 t-tau (pg/ml) 38 (13.4-129.5) 37 (17.9-142.2) 0.686 p-tau (pg/ml) 32 (15.4-85.8) 31 (15.6-106.1) 0.198 Aβ42 (pg/ml) 1230 (276.8-2685.2) 1195 (265.2-3363.2) 0.500 Aβ40 (pg/ml) 8089 (2882.4-15146.0) 7671 (1275.3-16450.8) 0.369

Changes of CSF α-syn species and disease progression

All patients

The correlation between the changes of CSF biomarkers and PD progression over the period of follow-up

in the DATATOP cohort is shown in Table 3. A significant positive correlation was observed between the

changes in CSF levels of both t-α-syn and o-α-syn with the two-year follow-up time (rs = 0,28, p < 0.01 and

rs = 0,32, p < 0.01, respectively, Fig. 1A, B). The correlation between o-/t-α-syn ratio and UPDRS motor

change (r=-0.22, p<0.05) was significant also after adjustment for baseline UPDRS motor and follow-up

on the regression model (β=-0.09, p<0.05). We have found also a significant association between o-/t-α-

syn ratio and UPDRS axial change (r=-0.31, p<0.01) that was significant also in the regression model (β=-

0.03, p<0.001) and a correlation between p-tau and change in ADL (r=-0.20, p<0.05), but this was not

significant after adjustment for follow-up, baseline ADL and medications (β=-0.08, p=0.114). A strong

positive correlation between the changes in t-α-syn and o-α-syn was also noted (r= 0.84, p< 0.0001, Fig.1C).

Tremor dominant patients

We found a significant association between change of CSF p-tau levels and UPDRS motor change in tremor

dominant PD (r=-0.35, p<0.05), and this was significant also after adjustment (β=-0.44, p<0.05).

Correlation between changes of CSF biomarkers levels and clinical parameters in tremor dominant group

are presented in Supplementary Table 1.

PIGD

Interestingly the correlation between o-/t-α-syn ratio and UPDRS motor was stronger in PIGD PD (r=-0.41,

p<0.01) while it was not significant in the tremor dominant subtype (r=-0.05, p=0.782); the association

between o-/t-α-syn ratio and UPDRS motor in PIGD holds also after adjustment for follow-up and baseline

  109  

UPDRS motor (β=-0.14, p<0.05). Furthermore, we found a significant correlation between change of o-/t-

α-syn ratio and ADL (r=-0.30, p<0.05) and this was not significant in the multivariate model accounting

for the effect of follow-up time, baseline ADL and medications (β=-0.04, p=0.081). UPDRS axial was

correlated with o-/t-α-syn ratio (r=-0.34, p<0.05) and this was significant also after adjustment for baseline

UPDRS axial and follow-up time (β=-0.03, p<0.01). UPDRS axial was correlated with t-tau (r=0.31,

p<0.05) but this was not significant in the multivariate model (β=0.05, p=0.076). Correlation between

changes of CSF biomarkers levels and clinical parameters in PIGD group are presented in Supplementary

Table 2.

Intermediate

No significant association was noted between the change in CSF biomarkers and the clinical parameters

(see Supplementary Table 3).

Figure 1. Scatter plots indicating the correlation between changes in CSF t-, o-α-syn and o-/t-α-syn levels with follow-up time and changes in clinical staging.

The correlation between the change of CSF t- and o-α-syn with length of follow-up time (r =0·28, p < 0·01 and r =0·32, p < 0·01, respectively) (A-B). The correlation between the changes in CSF t-α-syn and o-α-syn over the two-year DATATOP study period (r= 0·84, p < 0·0001) (C) The dotted line is the 95% prediction interval for the calculated regression line (solid line).

  110  

Correlation of CSF α-syn species with AD biomarkers

The correlation between changes of CSF biomarkers levels are all shown in Table 4. Interestingly, a

significant correlation was found between the change of p-tau with the change of o-α-syn (r=-0.21, p<0.05)

and the change of o-/t-α-syn ratio (r=-0.23, p<0.05). Furthermore, the change of Aβ42 correlated with the

change of p-S129-/t-α-syn ratio (r=0.19, p<0.05), whereas, the change of Aβ40 correlated with change of

o-α-syn (r=-0.25, p<0.05), o-/t-α-syn ratio (r=-0.23, p<0.05) and p-S129-/t-α-syn ratio (r=0.21, p<0.05).

We have also investigated the correlations between baseline levels of CSF biomarkers (Supplementary

Table 4). T-tau correlated with o-α-syn (r=-0.21, p<0.05) and o-/t-α-synuclein ratio (r=-0.23, p<0.05),

whereas, p-tau correlated with o-α-syn (r=-0.22, p<0.05) and o-/t-α-syn ratio (r = -0.26, p<0.05). Moreover,

Aβ42 correlated with t-α-syn (r=-0.21, p<0.05), o-α-syn (r=-0.35, p<0.001) and o-/t-α-syn ratio (r=-0.24,

p<0.05). Similar findings were noted with Aβ40, which also correlated with t-α-syn (r=-0.21, p<0.05) o-α-

syn (r=-0.37, p<0.001) and o-/t-α-syn ratio (r=-0.27, p<0.05).

Table 3. Correlation between changes of CSF biomarkers levels and disease duration, follow-up

time, changes of clinical outcomes/staging in all patients. Spearman’s rho and p-values (*p<0.05,

**p<0.01).

Follow-up HY ADL UPDRS motor

UPDRS tremor

UPDRS rigidity

UPDRS bradykinesia

UPDRS Axial

t-α-syn 0.28** 0.07 0.03 0.04 -0.07 0.02 0.04 0.02 p-S129-α-syn 0.04 -0.09 -0.07 -0.07 0.06 -0.10 0.01 0.04 o-α-syn 0.32** 0.11 -0.01 0.02 -0.09 0.04 0.00 -0.07 t-tau -0.03 0.09 -0.08 0.01 0.01 -0.02 0.07 0.10 p-tau -0.05 -0.15 -0.20 -0.19 0.01 -0.15 -0.07 -0.04 Aβ42 0.01 -0.16 0.04 -0.05 0.13 -0.02 -0.07 0.07 Aβ40 -0.07 -0.04 0.10 0.01 0.10 0.06 0.02 0.15 p-S129-/t-α-syn -0.24* -0.02 0.02 0.02 0.17 -0.05 0.11 0.10 o-/t-α-syn 0.15 -0.08 -0.17 -0.22* -0.19 -0.01 -0.03 -0.31**

  111  

Table 4. Correlation between changes of CSF biomarkers levels (* p<0.05, ** p<0.01). t-α-syn p-S129-α-syn o-α-syn o-/t-α-syn p-S129-/t-α-syn t-tau p-tau Aβ42 Aβ40 t-α-syn 1.00 p-S129-α-syn -0.29** 1.00 o- α -syn 0.84*** -0.24* 1.00 o-/t- α-syn -0.02 1.00 p-S129-/t-α-syn -0.66*** -0.01 1.00 t-tau 0.02 -0.09 0.01 -0.12 -0.07 1.00 p-tau -0.12 0.07 -0.21* -0.23* 0.09 0.51*** 1.00 Aβ42 -0.12 0.17 -0.17 -0.16 0.19* 0.14 0.22* 1.00 Aβ40 -0.18 0.12 -0.25** -0.23* 0.21* 0.17 0.28** 0.78*** 1.00

DISCUSSION CSF biomarker analyses and longitudinal clinical evaluations were conducted in a large cohort of early

diagnosed PD patients who participated in the DATATOP study. In the two-year DATATOP follow-up

period, CSF t- and o-α-syn levels were significantly higher, while p-S129-α-syn levels were significantly

lower, at follow-up compared to baseline. The changes in t- and o-α-syn levels, but not p-S129-α-syn levels,

positively correlated with follow-up time, further emphasizing the longitudinal increase in CSF t- and o-α-

syn levels over disease progression. Our findings are in disagreement with those reported recently[21], in

contrast these authors observed no significant longitudinal change in CSF t-α-syn, whereas, CSF p-S129-

α-syn increased longitudinally[21]. In a recent study by Shaw and colleagues [22], cross-sectional analysis

of the longitudinal PPMI study showed a significant reduction of CSF t-α-syn in PD compared to controls.

In contrast, CSF t-α-syn levels showed a longitudinal increase over the two-year DATATOP follow-up

period. However, the lack of control subjects in the DATATOP study, the small window of follow-up time,

the use of different quantification methods and the impact of pre-analytical confounding factors (types of

tubes, spinning conditions, storage conditions, etc.), hinder the comparison between both studies.

While the change in CSF t- or o-α-syn levels alone did not correlate with the longitudinal motor progression

of the disease in our DATATOP cohort, the change in o-/t-α-syn ratio significantly associated with UPDRS

motor scores. More interestingly, sub-group analysis of PD patients revealed a stronger correlation between

the change in o-/t-α-syn ratio and the change in UPDRS motor in PIGD group, whereas, the same correlation

was absent in tremor dominant group.

To our knowledge, this is the first study coupling the CSF biochemical profile of α-syn species with the

clinical PD phenotype. In this field, it is interesting to mention a recent functional MRI study measuring

  112  

brain activity in which different brain activation patterns could be found in tremor-dominant PD patients as

compared to those nontremor-dominant [23]. Our data seem to suggest that the PD clinical phenotypes

could be characterized by different CSF patterns. This finding, if confirmed in other series, might have

relevant implication in diagnosis, prognosis and treatment of PD. These results together with our previous

studies provide strong evidence for the important role of o-α-syn in the pathogenesis of PD. The correlations

noted between α-syn species and AD biomarkers, although weak, could also hold a potential in tracking PD

progression.

The PD subjects in the DATATOP study were at early stage of the disease, unlike most other clinical trials.

At baseline, these PD patients had minimal disability and were followed until just prior to the initiation of

the dopamine replacement therapy, which is reflected in their low H&Y scores. The fact that the patients

were at an early stage of disease with limited motor disability may explain the lack of an association

between the baseline or follow-up CSF biomarkers and disease severity. We did observe significant changes

of CSF α-syn levels over the two-year follow-up period, however, it is likely that a correlation between

CSF α-syn levels and clinical parameters will become more apparent with a longer follow-up. This will

require large-scale, prospective and well-controlled studies, especially those that include PD subjects with

neuroimaging and healthy controls such as the on-going Parkinson’s Progression Markers Initiative.

To our knowledge, this is the first study that combines measurements of different species of α-syn and AD

biomarkers and examined their longitudinal changes. In this study we demonstrated that quantification of

α-syn species in CSF has strong potential for tracking PD progression. However, we have to interpret our

findings with cautious since the DATATOP cohort is subject to several limitations; 1) the CSF samples

from the DATATOP study were stored for very long time (>25 years), and long storage could lead to

degradation, aggregation/precipitation and/or change in the phosphorylation state of α-syn; 2) the collection

and the process procedures of the CSF samples were not standardized; 3) and the absence of healthy controls

in the DATATOP study. Nevertheless, these caveats don’t underrate the importance of our original findings,

especially in the aspect of PD progression. Our interesting findings still need to be further validated in large-

scale, prospective and well-controlled studies, especially those that include subjects with neuroimaging-

supported definite PD and healthy controls and where CSF samples are collected and processed using

rigorous standardization as in the ongoing Parkinson’s Progression Markers Initiative.

Altogether, our current study shows that CSF α-syn species have a dynamic pattern along the course of the

disease, suggesting their possible role as progression biomarkers for PD.

  113  

REFERENCES

[1]   J.  Jankovic,  "Parkinson's  disease:  clinical  features  and  diagnosis,"  (in  eng),  J  Neurol  Neurosurg  Psychiatry,  vol.  79,  no.  4,  pp.  368-­‐76,  Apr  2008.  

[2]   M.  G.  Spillantini,  M.  L.  Schmidt,  V.  M.  Lee,  J.  Q.  Trojanowski,  R.  Jakes,  and  M.  Goedert,  "Alpha-­‐synuclein  in  Lewy  bodies,"  (in  eng),  Nature,  vol.  388,  no.  6645,  pp.  839-­‐40,  Aug  28  1997.  

[3]   K.  E.  Paleologou,  G.  B.  Irvine,  and  O.  M.  El-­‐Agnaf,  "Alpha-­‐synuclein  aggregation  in  neurodegenerative  diseases  and  its  inhibition  as  a  potential  therapeutic  strategy,"  (in  eng),  Biochem  Soc  Trans,  vol.  33,  no.  Pt  5,  pp.  1106-­‐10,  Nov  2005.  

[4]   O.  M.  El-­‐Agnaf  et  al.,  "Aggregates  from  mutant  and  wild-­‐type  alpha-­‐synuclein  proteins  and  NAC  peptide  induce  apoptotic  cell  death  in  human  neuroblastoma  cells  by  formation  of  beta-­‐sheet  and  amyloid-­‐like  filaments,"  (in  eng),  FEBS  Lett,  vol.  440,  no.  1-­‐2,  pp.  71-­‐5,  Nov  1998.  

[5]   O.  M.  el-­‐Agnaf,  G.  B.  Irvine,  and  D.  J.  Guthrie,  "Conformations  of  beta-­‐amyloid  in  solution,"  (in  eng),  J  Neurochem,  vol.  68,  no.  1,  pp.  437-­‐9,  Jan  1997.  

[6]   T.  Tokuda  et  al.,  "Decreased  alpha-­‐synuclein  in  cerebrospinal  fluid  of  aged  individuals  and  subjects  with  Parkinson's  disease,"  (in  eng),  Biochem  Biophys  Res  Commun,  vol.  349,  no.  1,  pp.  162-­‐6,  Oct  2006.  

[7]   L.  Parnetti  et  al.,  "Differential  role  of  CSF  alpha-­‐synuclein  species,  tau,  and  Aβ42  in  Parkinson's  Disease,"  (in  eng),  Front  Aging  Neurosci,  vol.  6,  p.  53,  2014.  

[8]   K.  D.  van  Dijk,  M.  Bidinosti,  A.  Weiss,  P.  Raijmakers,  H.  W.  Berendse,  and  W.  D.  van  de  Berg,  "Reduced  α-­‐synuclein  levels  in  cerebrospinal  fluid  in  Parkinson's  disease  are  unrelated  to  clinical  and  imaging  measures  of  disease  severity,"  (in  eng),  Eur  J  Neurol,  vol.  21,  no.  3,  pp.  388-­‐94,  Mar  2014.  

[9]   L.  Parnetti  et  al.,  "Cerebrospinal  fluid  biomarkers  in  Parkinson  disease,"  (in  eng),  Nat  Rev  Neurol,  vol.  9,  no.  3,  pp.  131-­‐40,  Mar  2013.  

[10]   L.  Parnetti  et  al.,  "Cerebrospinal  fluid  Tau/α-­‐synuclein  ratio  in  Parkinson's  disease  and  degenerative  dementias,"  (in  eng),  Mov  Disord,  vol.  26,  no.  8,  pp.  1428-­‐35,  Jul  2011.  

[11]   B.  Mollenhauer,  J.  J.  Locascio,  W.  Schulz-­‐Schaeffer,  F.  Sixel-­‐Döring,  C.  Trenkwalder,  and  M.  G.  Schlossmacher,  "α-­‐Synuclein  and  tau  concentrations  in  cerebrospinal  fluid  of  patients  presenting  with  parkinsonism:  a  cohort  study,"  (in  eng),  Lancet  Neurol,  vol.  10,  no.  3,  pp.  230-­‐40,  Mar  2011.  

[12]   N.  K.  Majbour  et  al.,  "Oligomeric  and  phosphorylated  alpha-­‐synuclein  as  potential  CSF  biomarkers  for  Parkinson's  disease,"  (in  eng),  Mol  Neurodegener,  vol.  11,  no.  1,  p.  7,  2016.  

[13]   M.  Helwig  et  al.,  "Brain  propagation  of  transduced  α-­‐synuclein  involves  non-­‐fibrillar  protein  species  and  is  enhanced  in  α-­‐synuclein  null  mice,"  (in  ENG),  Brain,  Dec  2015.  

[14]   M.  T.  Ardah  et  al.,  "Structure  activity  relationship  of  phenolic  acid  inhibitors  of  α-­‐synuclein  fibril  formation  and  toxicity,"  (in  eng),  Front  Aging  Neurosci,  vol.  6,  p.  197,  2014.  

[15]   M.  T.  Ardah  et  al.,  "Ginsenoside  Rb1  inhibits  fibrillation  and  toxicity  of  alpha-­‐synuclein  and  disaggregates  preformed  fibrils,"  (in  eng),  Neurobiol  Dis,  vol.  74,  pp.  89-­‐101,  Feb  2015.  

  114  

[16]   L.  M.  Shaw  et  al.,  "Qualification  of  the  analytical  and  clinical  performance  of  CSF  biomarker  analyses  in  ADNI,"  (in  eng),  Acta  Neuropathol,  vol.  121,  no.  5,  pp.  597-­‐609,  May  2011.  

[17]   "DATATOP:  a  multicenter  controlled  clinical  trial  in  early  Parkinson's  disease.  Parkinson  Study  Group,"  (in  eng),  Arch  Neurol,  vol.  46,  no.  10,  pp.  1052-­‐60,  Oct  1989.  

[18]   J.  Jankovic  et  al.,  "Variable  expression  of  Parkinson's  disease:  a  base-­‐line  analysis  of  the  DATATOP  cohort.  The  Parkinson  Study  Group,"  (in  eng),  Neurology,  vol.  40,  no.  10,  pp.  1529-­‐34,  Oct  1990.  

[19]   M.  E.  Domellöf,  E.  Elgh,  and  L.  Forsgren,  "The  relation  between  cognition  and  motor  dysfunction  in  drug-­‐naive  newly  diagnosed  patients  with  Parkinson's  disease,"  (in  eng),  Mov  Disord,  vol.  26,  no.  12,  pp.  2183-­‐9,  Oct  2011.  

[20]   N.  N.  Vaikath  et  al.,  "Generation  and  characterization  of  novel  conformation-­‐specific  monoclonal  antibodies  for  α-­‐synuclein  pathology,"  (in  eng),  Neurobiol  Dis,  vol.  79,  pp.  81-­‐99,  Jul  2015.  

[21]   T.  Stewart  et  al.,  "Phosphorylated  α-­‐synuclein  in  Parkinson's  disease:  correlation  depends  on  disease  severity,"  (in  eng),  Acta  Neuropathol  Commun,  vol.  3,  p.  7,  2015.  

[22]   J.  H.  Kang  et  al.,  "Association  of  cerebrospinal  fluid  β-­‐amyloid  1-­‐42,  T-­‐tau,  P-­‐tau181,  and  α-­‐synuclein  levels  with  clinical  features  of  drug-­‐naive  patients  with  early  Parkinson  disease,"  (in  eng),  JAMA  Neurol,  vol.  70,  no.  10,  pp.  1277-­‐87,  Oct  2013.  

[23]   J.  Prodoehl,  P.  J.  Planetta,  A.  S.  Kurani,  C.  L.  Comella,  D.  M.  Corcos,  and  D.  E.  Vaillancourt,  "Differences  in  brain  activation  between  tremor-­‐  and  nontremor-­‐dominant  Parkinson  disease,"  (in  eng),  JAMA  Neurol,  vol.  70,  no.  1,  pp.  100-­‐6,  Jan  2013.  

  115  

                                                                 

  116    

!!

CHAPTER 5 Increased Levels of CSF Total but not Oligomeric or Phosphorylated Forms of Alpha-Synuclein in Patients Diagnosed with Probable Alzheimer’s Disease

  117  

CHAPTER 5 | INCREASED LEVELS OF CSF TOTAL BUT NOT OLIGOMERIC OR PHOSPHORYLATED FORMS OF ALPHA-SYNUCLEIN IN PATIENTS DIAGNOSED WITH PROBABLE ALZHEIMER’S DISEASE

Nour K. Majbour, MSc1, 2†, Davide Chiasserini, PhD3†, Nishant N. Vaikath, MSc1, 4, Paolo Eusebi, PhD3,

Takahiko Tokuda, MD, PhD5, Wilma van de Berg, PhD2, Lucilla Parnetti, MD, PhD3, Paolo Calabresi,

MD3, 6, Omar M.A. El-Agnaf, PhD1,7 1Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 5825, Doha, Qatar 2Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU University Medical Centre, Amsterdam, The Netherlands. 3Dipartimento di Medicina, sezione di Neurologia, Università degli Studi di Perugia, Perugia, Italy 4Neural Plasticity and Repair Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, BMC A10, Lund University, Lund, Sweden 5Department of Neurology, Research Institute for Geriatrics, Kyoto Prefectural University of Medicine, Kyoto, 602-0841, Japan 6IRCCS Fondazione S. Lucia, Roma, Italy 7Life Sciences Division, College of Science and Engineering, Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, PO Box 5825, Doha, Qatar †These authors contributed equally to this work.

Sci Rep. 2017 Jan 10;7:40263. doi: 10.1038/srep40263.

  118  

Abstract Several studies reported an association between CSF alpha-synuclein (α-syn) and tau in Alzheimer’s

disease (AD), and demonstrated the significance of α-syn in improving the diagnostic sensitivity/specificity

of classical AD CSF biomarkers. In the current study, we measured CSF levels of different α-syn species

in a cohort of AD patients (n=225) who showed a CSF profile typical of AD at baseline as well as in

cognitively intact controls (n=68). CSF total α-syn (t-α-syn) significantly increased in the AD group (p

<0.0001) compared to controls, while oligomeric- and phosphorylated-Ser129-α-syn did not change

significantly. ROC analysis showed a sensitivity of 85% and a specificity of 84% (AUC = 0.88) in

distinguishing AD from controls. T-α-syn levels correlated positively with tau species in AD group and

negatively with baseline MMSE score. Our data support the added value of measurement of CSF α-syn

species for further characterization of the CSF AD profile.

Introduction α-Synuclein (α-syn) is a pre-synaptic neuronal protein that has been linked to a number of

neurodegenerative disorders named as “synucleinopathies” [1]. However, the role of α-syn in the

pathogenesis of Alzheimer’s disease (AD) has been increasingly recognized since Uéda et al. reported the

presence of a non-Aβ component in the extracellular plaques found in the brains of AD patients, which was

shown to be a fragment of α-syn [2]. In fact, the role of α-syn became of particular interest when the co-

existence of α-syn and tau pathology was observed in the brains of patients with AD, Parkinson’s disease

(PD) and dementia with Lewy bodies (DLB) [3]. Although total tau (t-tau), phosphorylated tau (p-tau) and

amyloid beta-42 (Aβ42) are state biomarkers of AD, some of the neuropathological changes observed in

the brains of AD patients are not captured by these traditional biomarkers, such as α-syn inclusions “LBs”.

Therefore, there is a need for additional biomarkers that can lead to better understanding and differential

diagnosis. The aim of our study was to measure the levels of CSF α-syn species total- (t-), oligomeric- (o-

) and phosphorylated-Ser129- (p-S129-) α-syn in a cohort of patients clinically diagnosed with probable

AD who also exhibited a CSF profile positive for AD biomarkers, in support of the clinical diagnosis. To

evaluate the diagnostic performance of CSF α-syn species in AD patients, we also included a control group

composed of subjects who did not exhibit cognitive decline and who displayed CSF profiles negative for

AD biomarkers. These were patients who had been diagnosed with other neurological diseases (OND).

The correlation of CSF α-syn species with t-tau, p-tau and Aβ42 was also investigated.

Methods Patients and CSF sampling

The AD cohort enrolled in this study included patients who attended the Center for Memory Disturbances

of the University of Perugia between 2008 and 2011 and underwent CSF analysis for diagnostic purposes

  119  

after informed written consent. All of the patients underwent the following: a thorough clinical examination

by experienced neurologists; a neuropsychological assessment, including the Mini Mental State

Examination (MMSE); analysis of blood chemistry; and a brain CT and/or MRI scan to exclude major

cerebrovascular diseases or other pathological brain conditions (e.g., hydrocephalus, tumors, hematomas,

abscesses, etc.). The AD group was composed of 225 patients diagnosed with probable AD according to

the research criteria proposed by Dubois et al.[4] (M/F = 96/129). Each patient also had a CSF biomarker

profile indicative of AD, according to the cut-offs used in our center (Aβ42 < 800, t-tau > 300 and p-tau >

60). As neurological controls, we selected 68 subjects who were admitted to the neurological ward and

underwent CSF tapping for diagnostic reasons (e.g., headache, suspected myelopathy, etc.), were without

clinical evidence of cognitive impairment and had CSF biomarkers profile negative for AD (other

neurological diseases group, OND). In all subjects enrolled in this study, lumbar puncture was performed

as a routine diagnostic procedure between 8:00 and 10:00 a.m. CSF (10 mL) was collected in sterile

polypropylene tubes, centrifuged for 10 minutes at 2000 x g, divided into 0.5 mL aliquots and immediately

frozen at -80°C. CSF samples were collected according to a standard protocol following international

guidelines[5].The study was approved by the local Ethical Committee (Comitato Etico delle Aziende

Sanitarie della Regione Umbria - CEAS Umbria) and informed written consent was signed by all patients

enrolled or by their legal representatives. The work was carried out according to the Declaration of Helsinki.

Immunoassays to quantify α-syn species in the CSF

CSF t-, o- and p-S129-α-syn levels were measured using our recently published ELISA assays [6]. Briefly,

for measuring t-α-syn, a 384-well ELISA microplate was coated by overnight incubation at 4°C with 0.1

µg/ml Syn-140 (sheep anti-α-syn polyclonal antibody) in 200 mM NaHCO3, pH 9.6 (50 µl/well). Similarly,

Syn-140 was used for measuring p-S129-α-syn, while the conformation-specific monoclonal antibody

(Syn-O2) [7], which is specific for α-syn oligomers (0.2 µg/ml), was used as the primary antibody to capture

o-α-syn. The plate was then washed with phosphate-buffered saline containing 0.05% Tween-20 (PBST)

and incubated with 100 µl/well of blocking buffer (PBST containing 2.5% gelatin) for 2 hours at 37°C.

After washing, 50 µl of the CSF samples (thawed on ice and Tween-20 added to a final concentration of

0.05%) were added to each well, and the plate was incubated at 37°C for another 2.5 hours. Antibodies

included 11D12 (mouse anti-α-syn monoclonal antibody) for measuring t-α-syn, PS129 (mouse anti-pS129-

α-syn monoclonal antibody) for measuring pS129-α-syn and FL-140 (rabbit polyclonal antibody, Santa

Cruz Biotechnology, Santa Cruz, CA, USA) for measuring o-α-syn were diluted to the desired

concentration (1:5000, 1:1,000 and 1:1,000, respectively) in the blocking buffer before being added to the

corresponding wells and incubated at 37°C for 2 hours. Next, the plate was washed and then incubated for

2 hours at 37°C with 50 µl/well of species-appropriate secondary antibodies: donkey anti-mouse IgG HRP

or goat anti-rabbit IgG HRP (Jackson ImmunoResearch, US), diluted in blocking buffer (1:20,000). After

  120  

washing, the plate was incubated with 50 µl/well of an enhanced chemiluminescent substrate (SuperSignal

ELISA Femto, Pierce Biotechnology, Rockford, IL). Then, the chemiluminescence (relative light units)

was immediately measured using a VICTOR™ X3 multilabel plate reader (PerkinElmer). The standard

curve for the ELISA assays was carried out using 50 µl/well of serial dilutions of recombinant human α-

syn, p-S129-α-syn or o-α-syn in artificial CSF. The samples were screened in a blinded fashion and tested

randomly. All the results were confirmed with at least two independent experiments. A series of internal

controls was also run to check for run-to-run variations.

Measurement of AD biomarkers

CSF Aβ42, total tau, and p-tau were measured using an ELISA technique (INNOTEST ß amyloid 1-42,

hTAU-Ag, p-TAU 181 Ag, Fujirebio Europe, Gent, Belgium) as previously described [8].

Data analysis and statistics

Statistical analysis was performed using R software v. 2.15. Continuous variables were described by the

median and interquartile range because data distributions were skewed. Correlations were calculated using

Spearman’s Rho (rs). The Mann-Whitney test was used for initial comparisons between the two diagnostic

groups (p < 0.05). The accuracy of the diagnostic value of the biomarkers [9] was assessed by calculating

the area under the curve (AUC) of the receiver operating characteristic (ROC) curve [10]. Cut-off values

were calculated using sensitivity and specificity values that maximized Youden’s index. Because there was

a significant difference in age (p <0.0001) and in the distribution of gender (p <0.01) between the groups,

we corrected for these using a logistic regression approach to adjust for covariates. All CSF samples with

an erythrocyte count > 500 cells/µl were excluded from further analysis, as traces of blood may influence

CSF α-syn levels [11, 12].

Results

Patients’ characteristics

Demographic data and clinical features are reported in Table 1. The AD group had median disease duration

of 2 years at the time of the lumbar puncture. No significant difference in the number of years of education

was present. As expected, baseline MMSE scores were significantly lower in the AD group (p < 0.0001).

  121  

Table 1. Demographic data and clinical features of OND and AD subjects.

Demographics OND AD

p-value (n=68) (n=225)

Sex (M) 42 (62%) 96 (43%) 0.0094

Age 63.7 ± 10.6 71.2 ± 8.8 <0.0001

Education 9.1 ± 5.1 8.2 ± 4.5 0.3895

Disease years - 2.5 ± 1.4 - MMSE 26.1 ± 3.2 21.1 ± 3.2 <0.0001

Levels of α-syn species in the CSF of AD patients and control subjects

Due to the differences between the groups in some demographic and clinical parameters we used a logistic

regression approach to assess the diagnostic performance of the α-syn species, adjusting for covariates such

as sex, age and MMSE (Table 2). CSF t-α-syn levels were significantly higher in the AD group compared

to the OND group (p <0.0001, Fig. 1A), whereas CSF o-α-syn and p-S129-α-syn levels were similar in both

diagnostic groups (p = 0.22 and p = 0.35, respectively, Fig. 1B, C). As a consequence, the o-/t-α-syn ratio

and p-S129-/t-α-syn ratio were significantly lower in AD subjects compared to OND subjects (p < 0.001

and p < 0.0001 respectively, Fig. 1D, E).

Table 2. P-values of OND vs. AD comparisons of CSF biomarkers levels after adjustment for covariates (age, gender) using a regression model.

Biomarker OND AD

p-values (n=68) (n=225)

t-tau 202.5 ± 55.5 776.1 ± 476.2 <0.0001 p-tau 42.1 ± 9.5 105.5 ± 46.7 <0.0001 t-α-syn (pg/ml) 1119.0 ± 1114.4 2200.0 ± 1189.9 <0.0001 p-S129-α-syn (pg/ml) 81.8 ± 55.0 97.5 ± 61.8 0.346 o-α-syn (pg/ml) 158.5 ± 79.8 162.5 ± 113.3 0.2291 o-/t-α-syn 0.17 ± 0.08 0.09 ± 0.08 0.0003 p-S129-/t-α-syn 0.08 ± 0.06 0.04 ± 0.05 <0.0001

  122  

Figure 1. Boxplots of the values of CSF biomarkers observed in the AD and OND groups.

Individual values of the CSF levels of (A) t-α-syn, (B) o-α-syn, and (C) p-S129-α-syn and the (D) o-/t-α-syn ratio and (E) p-S129-/t-α-syn ratio in patients with AD and OND. Horizontal bold lines indicate the medians; upper and lower horizontal lines indicate the range of values.

ROC analysis was carried out for the α-syn species (Fig. 2), and t-α-syn achieved the best diagnostic

performance compared to o-α-syn or p-S129-α-syn, with a sensitivity of 85% and a specificity of 84%

(AUC = 0.87, 95% CI=0.82-0.93, and cut-off =1260 pg/ml). However, neither the o-/t-α-syn ratio nor the

p-S129-/t-α-syn ratio improved the global diagnostic performance of t-α-syn in detecting AD (sensitivity =

87%, specificity = 79%, AUC = 0.82, 95% CI=0.76-0.87, and cut-off = 11.5; sensitivity=50%,

specificity=84%, AUC= 0.71, 95% CI= 0.63-0.78, and cut-off= 8.0, respectively).

  123  

Figure 2. Receiver Operating Curves Analysis

ROC curves of t-α-syn, o-α syn, p-S129-α-syn, the o-/t-α-syn ratio and the p-S129-/t-α-syn ratio and the fitted values of the multivariable logistic regression model.

Correlation of CSF α-syn species with AD biomarkers and clinical features

Correlation analysis of all the CSF biomarkers in the diagnostic groups is reported in Supplementary Table

1. As previously reported [13], t-tau and p-tau CSF levels were significantly correlated in both diagnostic

groups; however, the positive correlation was much stronger in the AD group. Interestingly, a significant

positive correlation between t-α-syn and tau species was only noted in the AD group (r =0.31, p < 0.001 for

t-tau; r = 0.30, p < 0.001 for p-tau, Fig. 3A, B). Although similar correlations were observed within the

OND group, they did not reach statistical significance. The levels of CSF o-α-syn positively correlated with

t-α-syn levels (r =0.20, p < 0.01). On the other hand, in both the AD and OND groups, no significant

correlations between o- or p-S129-α-syn and AD CSF biomarkers were observed. The correlation analysis

between the CSF biomarkers and clinical parameters in the AD and OND groups is reported in

Supplementary Table 2. As expected, both t-tau and p-tau were negatively correlated with MMSE scores

in the AD group. In addition, a significant inverse correlation between CSF t-α-syn levels and MMSE

baseline scores was noted only within the AD group (r = -0.21, p <0.01, Fig. 3C). No significant correlations

  124  

were observed between the other α-syn species and age, education or disease duration in either diagnostic

group.

Figure 3. Scatter plots indicating the correlation between t-α-syn and t-tau, p-tau and MMSE scores.

ROC curves of t-α-syn, o-α syn, p-S129-α-syn, the o-/t-α-syn ratio and the p-S129-/t-α-syn ratio and the fitted values of the multivariable logistic regression model.

  125  

Discussion The role of α-syn as a putative AD biomarker has been investigated in several studies, largely with

contrasting results. Some studies have reported either unchanged or increased levels of t-α-syn in AD

patients compared to various control groups (healthy or other neurological conditions) [14-16].

In the present study, we used our newly developed, sensitive ELISAs to measure the concentration of t-α-

syn and other pathologically important CSF α-syn species (o- and p-S129-α-syn), which are mainly

associated with synucleinopathies. The CSF samples analyzed were selected from a large cohort of patients

clinically diagnosed with probable AD who also had an AD-positive CSF biomarkers profile. As a control

group, we included cognitively intact patients who had been diagnosed with other neurological diseases but

had an AD-negative CSF biomarkers profile. Our main findings were: i) a significant increase in CSF t-α-

syn levels in AD patients with respect to control subjects; ii) no significant change in CSF o- or p-S129-α-

syn levels in AD patients; and iii) a significant correlation of t-α-syn levels with tau species and MMSE in

the AD group.

The increase of t-α-syn in the AD group is in line with the findings of several previous reports [14, 17], but

contrasts with others [18, 19]. These discrepancies are likely due to several factors, including differences

in the assay platform used, lack of control of contamination of the samples with red blood cells and

heterogeneity of the patients included in the studies. In this study, we selected a cohort of AD patients by

not only relying on an accurate clinical and neuropsychological evaluation but also choosing the patients

according to the CSF profile of Aβ42, t-tau and p-tau, which further supports the initial AD diagnosis.

Whereas, the OND group included cognitively normal subjects with CSF biomarker profile negative for

AD, possibly excluding non-AD neurological disorders with neurodegeneration. This may explain the

difference in diagnostic performance among the studies. Korff and colleagues found a sensitivity of 65%

and a specificity of 74% in distinguishing AD patients from controls [20], while in the present work, the

performance of t-α-syn was better, reaching approximately 85% for both sensitivity and specificity.

A meaningful hypothesis about the increase of t-α-syn in the AD group relies on evidence of over-

expression in the brain tissue of AD patients, as previously shown [21], and/or on the neuronal damage

related to AD. This, in turn, can increase the release of α-syn from damaged cells into the brain’s interstitial

fluid and then into the CSF. This hypothesis is further supported by the significant positive correlation

between t-α-syn and tau species we found in our cohort. This correlation agrees with previous reports [17]

and was observed solely in the AD group. Tau protein is considered the prototypical biomarker of neuronal

damage because it is highly increased in the CSF of patients diagnosed with Creutzfeldt-Jakob disease [22],

a disease characterized by massive neurodegeneration, as well as in acute events such as traumatic brain

injury [23]. Notably, CSF α-syn is also increased in these conditions and in other neurological diseases in

which neurodegeneration is an important attribute [24-26], further stressing the parallelism between tau and

  126  

α-syn. In particular, the increase of α-syn in AD patients can be perceived as a marker of synapse loss and

synapse disruption. It is worth noting that, consistent with other studies [20, 27], we observed a significant

inverse correlation between the levels of CSF α-syn and cognitive function as measured by MMSE in our

AD group.

The unique role of α-syn in AD pathology relative to synucleinopathies is also supported by the lack of a

contribution of o- and p-S129-α-syn species to the diagnosis of AD. Substantial evidence suggests a

neurotoxic role for o-α-syn in the pathogenesis of synucleinopathies, both in vitro and in vivo [28], as α-

syn oligomers are increased in brain homogenates and the CSF of PD and DLB patients [29-31]. Here, we

found no significant differences in the level of o-α-syn in the AD group with respect to control subjects,

thus confirming our previous reports [30]. Likewise, phosphorylated α-syn has been previously linked to

PD pathogenesis, reflecting that most of the α-syn accumulated in LBs is phosphorylated at residue Ser129

[32, 33]. Elevated p-S129-/t-α-syn ratios have also been found in the CSF of PD patients [34]. In the current

study, p-S129-α-syn did not show any significant change in our AD cohort, supporting the hypothesis that

the oligomeric- and p-S129-α-syn species are intimately connected to the pathogenesis of

synucleinopathies.

One possible limitation of this study is the lack of neuropathological assessment. It has been shown that

AD biomarkers can detect the underlying AD pathology with high accuracy, but may not detect co-

morbidity caused by the presence of other protein aggregates (Toledo et al., Acta Neuropathol, 2012). Co-

morbidity may influence the accuracy of CSF biomarkers, including α-syn. Further studies should be

devoted to the understanding of the complex relationships between CSF AD biomarkers and α-syn species

across different types of dementia, possibly together with pathological confirmation. We also aim to include

subjects with α-syn related dementias such as DLB and PD with dementia in future research to broaden our

understanding of the potential role of α-syn as cognitive biomarkers.

In conclusion, our results in a well characterized cohort of AD patients and neurological controls further

support the notion that t-α-syn most likely represents a marker of neuron loss and/ or synaptic failure in

AD. Future studies should be aimed at understanding the added values of α-syn species to the core AD

biomarkers.

  127  

References

[1]   M.  G.  Spillantini  and  M.  Goedert,  "The  alpha-­‐synucleinopathies:  Parkinson's  disease,  dementia  with  Lewy  bodies,  and  multiple  system  atrophy,"  (in  eng),  Ann  N  Y  Acad  Sci,  vol.  920,  pp.  16-­‐27,  2000.  

[2]   K.  Uéda  et  al.,  "Molecular  cloning  of  cDNA  encoding  an  unrecognized  component  of  amyloid  in  Alzheimer  disease,"  (in  eng),  Proc  Natl  Acad  Sci  U  S  A,  vol.  90,  no.  23,  pp.  11282-­‐6,  Dec  1993.  

[3]   K.  Vekrellis,  M.  Xilouri,  E.  Emmanouilidou,  H.  J.  Rideout,  and  L.  Stefanis,  "Pathological  roles  of  α-­‐synuclein  in  neurological  disorders,"  (in  eng),  Lancet  Neurol,  vol.  10,  no.  11,  pp.  1015-­‐25,  Nov  2011.  

[4]   B.  Dubois  et  al.,  "Research  criteria  for  the  diagnosis  of  Alzheimer's  disease:  revising  the  NINCDS-­‐ADRDA  criteria,"  (in  eng),  Lancet  Neurol,  vol.  6,  no.  8,  pp.  734-­‐46,  Aug  2007.  

[5]   C.  E.  Teunissen  et  al.,  "A  consensus  protocol  for  the  standardization  of  cerebrospinal  fluid  collection  and  biobanking,"  Neurology,  vol.  73,  no.  22,  pp.  1914-­‐22,  Dec  1  2009.  

[6]   N.  K.  Majbour  et  al.,  "Oligomeric  and  phosphorylated  alpha-­‐synuclein  as  potential  CSF  biomarkers  for  Parkinson's  disease,"  (in  eng),  Mol  Neurodegener,  vol.  11,  no.  1,  p.  7,  2016.  

[7]   N.  N.  Vaikath  et  al.,  "Generation  and  characterization  of  novel  conformation-­‐specific  monoclonal  antibodies  for  α-­‐synuclein  pathology,"  (in  eng),  Neurobiol  Dis,  vol.  79,  pp.  81-­‐99,  Jul  2015.  

[8]   L.  Parnetti  et  al.,  "Performance  of  aβ1-­‐40,  aβ1-­‐42,  total  tau,  and  phosphorylated  tau  as  predictors  of  dementia  in  a  cohort  of  patients  with  mild  cognitive  impairment,"  (in  eng),  J  Alzheimers  Dis,  vol.  29,  no.  1,  pp.  229-­‐38,  2012.  

[9]   P.  Eusebi,  "Diagnostic  accuracy  measures,"  (in  eng),  Cerebrovasc  Dis,  vol.  36,  no.  4,  pp.  267-­‐72,  2013.  

[10]   X.  Robin  et  al.,  "pROC:  an  open-­‐source  package  for  R  and  S+  to  analyze  and  compare  ROC  curves,"  (in  eng),  BMC  Bioinformatics,  vol.  12,  p.  77,  2011.  

[11]   Z.  Hong  et  al.,  "DJ-­‐1  and  alpha-­‐synuclein  in  human  cerebrospinal  fluid  as  biomarkers  of  Parkinson's  disease,"  (in  eng),  Brain,  vol.  133,  no.  Pt  3,  pp.  713-­‐26,  Mar  2010.  

[12]   R.  Barbour  et  al.,  "Red  blood  cells  are  the  major  source  of  alpha-­‐synuclein  in  blood,"  (in  eng),  Neurodegener  Dis,  vol.  5,  no.  2,  pp.  55-­‐9,  2008.  

[13]   L.  Parnetti  et  al.,  "Cerebrospinal  fluid  Tau/α-­‐synuclein  ratio  in  Parkinson's  disease  and  degenerative  dementias,"  (in  eng),  Mov  Disord,  vol.  26,  no.  8,  pp.  1428-­‐35,  Jul  2011.  

[14]   J.  B.  Toledo,  A.  Korff,  L.  M.  Shaw,  J.  Q.  Trojanowski,  and  J.  Zhang,  "CSF  α-­‐synuclein  improves  diagnostic  and  prognostic  performance  of  CSF  tau  and  Aβ  in  Alzheimer's  disease,"  (in  eng),  Acta  Neuropathol,  vol.  126,  no.  5,  pp.  683-­‐97,  Nov  2013.  

[15]   K.  Kasuga  et  al.,  "Differential  levels  of  alpha-­‐synuclein,  beta-­‐amyloid42  and  tau  in  CSF  between  patients  with  dementia  with  Lewy  bodies  and  Alzheimer's  disease,"  (in  eng),  J  Neurol  Neurosurg  Psychiatry,  vol.  81,  no.  6,  pp.  608-­‐10,  Jun  2010.  

[16]   E.  Kapaki,  G.  P.  Paraskevas,  E.  Emmanouilidou,  and  K.  Vekrellis,  "The  diagnostic  value  of  CSF  α-­‐synuclein  in  the  differential  diagnosis  of  dementia  with  Lewy  bodies  vs.  normal  subjects  and  patients  with  Alzheimer's  disease,"  (in  eng),  PLoS  One,  vol.  8,  no.  11,  p.  e81654,  2013.  

[17]   S.  Slaets  et  al.,  "Increased  CSF  α-­‐synuclein  levels  in  Alzheimer's  disease:  Correlation  with  tau  levels,"  (in  ENG),  Alzheimers  Dement,  Jan  2014.  

[18]   F.  E.  Reesink  et  al.,  "CSF  alpha-­‐synuclein  does  not  discriminate  dementia  with  Lewy  bodies  from  Alzheimer's  disease,"  (in  eng),  J  Alzheimers  Dis,  vol.  22,  no.  1,  pp.  87-­‐95,  2010.  

[19]   A.  Ohrfelt  et  al.,  "Cerebrospinal  fluid  alpha-­‐synuclein  in  neurodegenerative  disorders-­‐a  marker  of  synapse  loss?,"  (in  eng),  Neurosci  Lett,  vol.  450,  no.  3,  pp.  332-­‐5,  Feb  2009.  

  128  

[20]   A.  Korff,  C.  Liu,  C.  Ginghina,  M.  Shi,  J.  Zhang,  and  A.  s.  D.  N.  Initiative,  "α-­‐Synuclein  in  cerebrospinal  fluid  of  Alzheimer's  disease  and  mild  cognitive  impairment,"  (in  eng),  J  Alzheimers  Dis,  vol.  36,  no.  4,  pp.  679-­‐88,  2013.  

[21]   M.  E.  Larson  et  al.,  "Soluble  α-­‐synuclein  is  a  novel  modulator  of  Alzheimer's  disease  pathophysiology,"  (in  eng),  J  Neurosci,  vol.  32,  no.  30,  pp.  10253-­‐66,  Jul  2012.  

[22]   T.  Skillbäck,  C.  Rosén,  F.  Asztely,  N.  Mattsson,  K.  Blennow,  and  H.  Zetterberg,  "Diagnostic  performance  of  cerebrospinal  fluid  total  tau  and  phosphorylated  tau  in  Creutzfeldt-­‐Jakob  disease:  results  from  the  Swedish  Mortality  Registry,"  (in  eng),  JAMA  Neurol,  vol.  71,  no.  4,  pp.  476-­‐83,  Apr  2014.  

[23]   S.  Magnoni  et  al.,  "Tau  elevations  in  the  brain  extracellular  space  correlate  with  reduced  amyloid-­‐β  levels  and  predict  adverse  clinical  outcomes  after  severe  traumatic  brain  injury,"  (in  eng),  Brain,  vol.  135,  no.  Pt  4,  pp.  1268-­‐80,  Apr  2012.  

[24]   K.  Blennow,  H.  Hampel,  M.  Weiner,  and  H.  Zetterberg,  "Cerebrospinal  fluid  and  plasma  biomarkers  in  Alzheimer  disease,"  (in  eng),  Nat  Rev  Neurol,  vol.  6,  no.  3,  pp.  131-­‐44,  Mar  2010.  

[25]   H.  Wang  et  al.,  "Cerebrospinal  fluid  α-­‐synuclein  levels  are  elevated  in  multiple  sclerosis  and  neuromyelitis  optica  patients  during  replase,"  (in  eng),  J  Neurochem,  vol.  122,  no.  1,  pp.  19-­‐23,  Jul  2012.  

[26]   T.  Kasai  et  al.,  "Increased  α-­‐synuclein  levels  in  the  cerebrospinal  fluid  of  patients  with  Creutzfeldt-­‐Jakob  disease,"  (in  eng),  J  Neurol,  vol.  261,  no.  6,  pp.  1203-­‐9,  Jun  2014.  

[27]   M.  E.  Larson  et  al.,  "Soluble  alpha-­‐synuclein  is  a  novel  modulator  of  Alzheimer's  disease  pathophysiology,"  in  J  Neurosci,  vol.  32no.  30)  United  States,  2012,  pp.  10253-­‐66.  

[28]   B.  Winner  et  al.,  "In  vivo  demonstration  that  alpha-­‐synuclein  oligomers  are  toxic,"  (in  eng),  Proc  Natl  Acad  Sci  U  S  A,  vol.  108,  no.  10,  pp.  4194-­‐9,  Mar  2011.  

[29]   T.  Tokuda  et  al.,  "Detection  of  elevated  levels  of  α-­‐synuclein  oligomers  in  CSF  from  patients  with  Parkinson  disease,"  (in  eng),  Neurology,  vol.  75,  no.  20,  pp.  1766-­‐72,  Nov  2010.  

[30]   O.  Hansson  et  al.,  "Levels  of  cerebrospinal  fluid  α-­‐synuclein  oligomers  are  increased  in  Parkinson's  disease  with  dementia  and  dementia  with  Lewy  bodies  compared  to  Alzheimer's  disease,"  (in  eng),  Alzheimers  Res  Ther,  vol.  6,  no.  3,  p.  25,  2014.  

[31]   K.  E.  Paleologou  et  al.,  "Detection  of  elevated  levels  of  soluble  alpha-­‐synuclein  oligomers  in  post-­‐mortem  brain  extracts  from  patients  with  dementia  with  Lewy  bodies,"  (in  eng),  Brain,  vol.  132,  no.  Pt  4,  pp.  1093-­‐101,  Apr  2009.  

[32]   J.  P.  Anderson  et  al.,  "Phosphorylation  of  Ser-­‐129  is  the  dominant  pathological  modification  of  alpha-­‐synuclein  in  familial  and  sporadic  Lewy  body  disease,"  (in  eng),  J  Biol  Chem,  vol.  281,  no.  40,  pp.  29739-­‐52,  Oct  2006.  

[33]   H.  Fujiwara  et  al.,  "alpha-­‐Synuclein  is  phosphorylated  in  synucleinopathy  lesions,"  (in  eng),  Nat  Cell  Biol,  vol.  4,  no.  2,  pp.  160-­‐4,  Feb  2002.  

[34]   Y.  Wang  et  al.,  "Phosphorylated  α-­‐synuclein  in  Parkinson's  disease,"  (in  eng),  Sci  Transl  Med,  vol.  4,  no.  121,  p.  121ra20,  Feb  2012.  

           

  129    

!!

CHAPTER 6 CSF Oligomeric Alpha-Synuclein Levels as A Preclinical Marker of Parkinson’s Disease: A Study in LRRK2 Mutation Carriers and Sporadic Parkinson’s Disease Cases

  130  

CHAPTER 6 |   CSF OLIGOMERIC ALPHA-SYNUCLEIN LEVELS AS A PRECLINICAL MARKER OF PARKINSON’S DISEASE: A STUDY IN LRRK2 MUTATION CARRIERS  Nour K. Majbour, MSc1,2, Jan O. Aasly, MD, PhD3,4, Eldbjørg Hustad, MD3,4, Mercy A. Thomas,

MSc1, Muneera Fayyad, MSc1, Nishant N. Vaikath, MSc1, Naser Elkum, PhD5, Wilma D. J. van

de Berg, PhD2, Takahiko Tokuda, MD, PhD6, Brit Mollenhauer, PhD7, Henk W. Berendse, MD,

PhD8, Omar M.A. El-Agnaf, PhD1

1Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad

Bin Khalifa University (HBKU), Education City, Qatar 2Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU University

Medical Centre, Amsterdam, The Netherlands 3Department of Neuroscience, Norwegian University of Science and Technology, (NTNU),

Trondheim, Norway 4Department of Neurology, St. Olav’s Hospital, University Hospital of Trondheim, Norway 5Clinical Epidemiology, Sidra Medical and Research Center, Doha, Qatar. 6Department of Neurology, Research Institute for Geriatrics, Kyoto Prefectural University of

Medicine, Kyoto, 602-0841 Japan 7Paracelsus-Elena Clinic, Centre of Parkinsonism and Movement Disorders, Department of

Neurology (BM) and Department of Neurosurgery (CT), Paracelsus-Elena Clinic, University

Medical Center Goettingen, Kassel, Germany.aracelsus-Elena-Klinik, Kassel, Germany 8Department of Neurology, Amsterdam UMC, location VU University Medical Centre,

Amsterdam, The Netherlands

Submitted to Neurology. Jan 2019

 

 

 

  131  

Abstract

Objective: To explore the diagnostic potential of alpha-synuclein (α-syn) oligomers in

cerebrospinal fluid (CSF) from individuals with leucine-rich repeat kinase 2 (LRRK2) mutation

carriers and subjects with sporadic Parkinson’s disease (PD).

Methods: We measured the levels of CSF total- (t-), oligomeric (o-) and phosphorylated S129

(pS129-) α-syn in symptomatic (n=21) and asymptomatic (n=50) LRRK2 mutation carriers,

subjects with the clinical diagnosis of PD (n=60) and age-matched healthy controls (HC) (n=39).

Results: CSF o-α-syn levels were significantly higher in sPD patients than in HC (P<0.001) and,

in asymptomatic LRRK2 mutation carriers than in HC (P=0.006). The area under the receiver

operating characteristic curve (AUC) indicated a sensitivity of 71.7% and specificity of 76.9%,

with an AUC of 0.76 with increased ratios of CSF o-/t-α-syn in sPD compared to HC. Similarly,

sensitivity and specificity rates didn’t exceed 80% for CSF o-/t-α-syn in symptomatic LRRK2

group. Assessing ROC curves of o-/t-α-syn ratio for pure sPD patients without any cognitive

impairment, generated greater sensitivity of 83.3%, and specificity of 90.6% with an AUC of 0.84.

Interestingly, asymptomatic LRRK2 mutations carriers scored moderate based on PET data,

presented with elevated levels of CSF t-α-syn levels, whereas, the subjects scored severe reported

high levels of CSF o-α-syn.

Conclusion: Our study indicates that CSF o-α-syn are potential surrogate biomarkers for early PD

detection. Moreover, our findings suggest that cognitive impairment in PD might be a certain

phenotype with a different aSyn biomarker profile, however, future biomarkers discovery studies

should consider stratifying the selected PD cohort by excluding patients with cognitive

impairment.

 

   

  132  

Introduction

Our understanding of the genetic basis of Parkinson’s disease (PD) has increased tremendously

over the past twenty years. Mutations in the gene encoding alpha-synuclein (α-syn) were the first

to be associated with genetic PD. Another monogenic causative factor in PD patients is leucine-

rich repeat kinase 2 (LRRK2); its role in PD occurrence is strongly supported by more than 100

LRRK2 variants.1 Asymptomatic carriers of LRRK2 mutations constitute an optimal population for

identifying preclinical biomarkers of PD for several reasons: 1) the long preclinical gap prior to

the onset of motor symptoms, 2) the reports of dopaminergic neuronal loss demonstrated by

positron emission tomography (PET) scanning, and 3) the similarity of these individuals’ clinical

phenotype to that of patients with sporadic PD (sPD). Several research groups, including ours,

reported a significant elevation of CSF o-α-syn and α-syn phosphorylated at Ser129 (pS129-α-syn)

in PD patients compared to the levels of these oligomers in controls, suggesting their potential role

as diagnostic biomarkers in the early detection of PD.2, 3 Here, we used our previously established

in-house-developed sandwich-based ELISAs,3 aiming to determine whether levels of CSF α-syn

species, including total (t-), o- and pS129-α-syn, allow the early detection of PD in presymptomatic

cases and are associated with the nigrostriatal dopaminergic deficit measured used PET and with

clinical parameters. We measured the levels of CSF t-, o- and pS129-α-syn species in a well-

characterized Norwegian cohort of 71 subjects with LRRK2 mutations: 21 symptomatic individuals

and 50 asymptomatic mutation carriers. In parallel, we included 60 patients with sPD and 39

healthy control (HC) subjects.

Methods

Patient selection and CSF sampling

Patient selection criteria and the method of CSF collection were extensively described in previous

publications.4, 5 In total, 72 Norwegian individuals from 12 different families with LRRK2

mutations were assessed in the current study. Twenty-one patients were clinically diagnosed with

PD, whereas 50 patients were healthy, asymptomatic LRRK2 mutation carriers when enrolled in

the study. These families have been extensively described in previous reports.4, 6, 7 In addition, 60

patients with sPD and 31 age-matched HC were recruited for this study the same as LRRK2

individuals from St. Olav’s Hospital at the University Hospital of Trondheim in Norway. PD was

diagnosed according to established clinical diagnostic criteria (UK Parkinson’s Disease Society  

  133  

Brain Bank). Disease stage was assessed according to the Hoehn and Yahr (H&Y) scale. All

patients with sPD were screened and confirmed to be negative for known LRRK2 mutations.

Patients with an age at onset of ≤50 years also tested negative for known pathogenic mutations in

Parkin and PINK1. All family members of LRRK2 patients were examined for clinical features of

PD by movement disorder specialists and found to be asymptomatic, although a few had mild

premotor signs and an increased Unified Parkinson’s Disease Rating Scale (UPDRS) score.8 The

LRRK2-mutant PD patients were taking levodopa, and some were taking dopamine agonists and/or

monoamine oxidase-B (MAO-B) inhibitors. Lumbar punctures were performed in overnight fasted

patients between 8 and 10 am. CSF samples were aliquoted in 1.2–1.5 ml low-binding tubes, and

one vial was sent for routine laboratory analysis (i.e., white and red blood cell count, total protein

and glucose levels, according to the Parkinson’s Progression Markers Initiative [PPMI] protocol),

whereas the majority of the vials were frozen fewer than 15 min after collection following

centrifugation at 2000 g at 4º C then subaliquoted and stored at -80 °C until further analysis. All

patients provided signed informed consent, and the study was approved by the Regional

Committee for Medical and Health Research Ethics.

PET scan analysis

Scanning was performed following scanning protocols as previously described.9 Within 1 year of

CSF sample collection, each subject was scanned with 3 different tracers, 18F-6-fluoro-L-dopa

(FD), as a marker for the uptake and decarboxylation of levodopa as well as the trapping of

dopamine in synaptic vesicles, 11C-(±) dihydrotetrabenazine (DTBZ), a vesicular monoamine

transporter 2 [VMAT2] ligand, and 11C-D-threomethylphenidate (MP), a dopamine transporter

[DAT] ligand. PET images were scored by the examiner as mild, moderate or severe.

Measurement of alpha-synuclein species via immunoassays

All immunoassays used to measure the different species of α-syn were developed in-house and

were thoroughly described in previous reports.3, 10, 11 Briefly, to capture t- or pS129-α-syn, a 384-

well ELISA microplate was coated with 0.1 or 0.5 µg/ml Syn-140 (a sheep anti-α-syn polyclonal

antibody) in 200 mM NaHCO3, pH 9.6 (50 µl/well) by overnight incubation at 4 °C, while 0.2

µg/ml of our mouse conformation-specific antibody, Syn-O2, was used to capture o-α-syn. After

incubation with 100 µl/well of blocking buffer (PBS-T containing 2.25% gelatin) for 2 h at 37 °C,

50 µl/well of the CSF samples (diluted 1:2 in artificial CSF) along with serial dilutions of

recombinant human t-, pS129-or o-α-syn (50 µl) were dispensed in each well, and the plate was

  134  

incubated at 37 °C for 2.5 h. After washing with PBS-T, 50 µl/well of 11D12 (a mouse anti-α-syn

monoclonal antibody), PS129 (a mouse anti-pS129-α-syn monoclonal antibody), or FL-140 (rabbit

polyclonal antibody, Santa Cruz Biotechnology, Santa Cruz, CA, USA), for measuring t-, o-, or

pS129-α-syn, respectively, were added to the corresponding wells and incubated at 37 °C for 2 h.

Next, the plates were washed and incubated for 2 h at 37 °C with 50 µl/well of species-appropriate

secondary antibody (goat anti-rabbit IgG HRP or donkey anti-mouse IgG HRP, Jackson

ImmunoResearch Laboratories Inc., USA, 1:20,000 dilution). After washing, the plates were

incubated with 50 µl/well of enhanced chemiluminescent substrate (Super Signal ELISA Femto,

Pierce Biotechnology, USA). The chemiluminescence, expressed in relative light units, was

immediately measured using a PerkinElmer Envision multilabel plate reader (PerkinElmer,

Finland). CSF samples were measured in a blinded fashion and randomized for analysis, with all

LRRK symptomatic/asymptomatic, PD and HC samples being tested together on the same ELISA

microplates. A series of internal controls was also run to check for run-to-run variations. The

concentrations of α-syn species in the samples were calculated using the corresponding standard

curves.

Statistical analysis

GraphPad Prism (GraphPad Prism Version 7.0a, GraphPad software, San Diego, CA) and IBM

SPSS software (version 24.0, Chicago, IL, USA) were used for the statistical analyses of the data.

Quantitative variables are expressed as the medians and interquartile ranges (IQRs) (25th and 75th

percentiles). Qualitative variables are expressed as counts or percentages and were analyzed by

Fisher’s exact test. The Mann-Whitney U test was used for comparisons among the diagnostic

groups. To evaluate the relationship between CSF α-syn species and the patients’ clinical data,

Spearman’s rho correlation analysis was used. To explore the potential diagnostic value of the

levels of CSF α-syn species and o- or pS129-/t-α-syn ratio in CSF, receiver operating characteristic

(ROC) curves were generated, and the area under the curve (AUC) was calculated. The cutoff

values were calculated using the sensitivity and specificity values that maximized Youden’s index

(sensitivity + specificity−1).  

 

 

  135  

Results

Patient population and demographics

The patient groups and demographic details are summarized in Table 1. Twenty-one of the 71

subjects (30%) with LRRK2 mutations analyzed had a manifest PD and were carrying either the

most common LRRK2 mutation, G2019S, or a different LRRK2 mutation, N1437H. These 21

subjects had a mean age of 58.0 ± 11 years. Fifty subjects of the 71 subjects (70%) were free of

symptoms of PD at the time of CSF sample collection. These asymptomatic LRRK2 mutation

carriers had a mean age of 57.2 ± 13.4 years. As comparison, we included samples from 60

manifest PD without genetic mutation. And 39 age-matched HCs were included in this study. No

significant difference was noted in disease duration between the symptomatic LRRK2 mutation

carriers and the PD patients. Moreover, no difference was found between the groups with regard

to the routine CSF levels of white cell count-red as well as the total protein, albumin, and glucose

levels, including the plasma glucose level. Controlling for age and sex did not significantly alter

any results.

Table 1| Demographics and CSF biomarkers by diagnostic group

 CSF alpha-synuclein levels

Comparisons of the CSF biomarker levels among the different diagnostic groups are summarized

in Table 2.

Table 2. a| CSF biomarkers among different diagnostic groups (Median and Interquartile range)

 

Table&1|&Demographics&and&CSF&biomarkers&by&diagnostic&groupHealthy(Controls

(n=(39)

sporadic(PD

(n=60)

LRRK2(asymptomatic(carriers

(n=(50)

LRRK2(symptomatic(carriers(

(n=(21)

Age((yrs) 50((18E80) 57((26E80) 57((27E83) 58((45E80)

Gender M=(21,(F=18 M=(34,(F=(26 M=(25,(F=(27 M=(4,(F=(17

Disease(Duration((yrs) N/A 3.5((1E6) N/A 7((5.3E20)

MoCA 27((27E29) 27((25E28) 27((26E28) 26((24E27)

UPDRSEIII N/A 23.5((19E28) 3((0.5E5) 23(((17E25)

HEY(grade N/A 2((2E2) 0((0EE0) 2((2E3)

*(Median(and(Interquartile(range(for(continious(values

Healthy(Controls(n=(39)

sporadic(PD(n=60)

LRRK2(asymptomatic(carriers(n=(50)

LRRK2(symptomatic(carriers((n=(21)

tB⍺Bsyn((ng/ml) 0.79((0.58B1.08) 0.57((0.47B0.70) 0.64((0.43B0.8) 0.61((0.35B0.80)oB⍺Bsyn((pg/ml) 161((148B186) 188((171B220) 182((157B220) 162((145B183)pS129B⍺Bsyn((pg/ml) 118((101B149) 139((114B163) 120((94B150) 122((108B156)oB/tB⍺Bsyn(% 22.6((15.4B27.4) 33.1((24.3B41.4) 31.3((21.6B45.9) 28.7((20.9B42.6)pS129B/tB⍺Bsyn(% 15((10.9B21.1) 25.4((16.4B30.2) 20.3((13B29.3) 24.4((18.1B29.7)

  136  

Table 2. b| CSF biomarkers among different diagnostic groups. One way ANOVA was used for

comparisons among all the groups, while Mann-Whitney U test was used for comparisons between

the two groups as illustrated in the table.

   

In comparison to HC (median and IQR = 0.79 (0.58-1.08) ng/ml, n = 39), levels of CSF t-α-syn

were significantly lower in the sPD (median and IQR = 0.57 (0.47-0.70) ng/ml, n = 60) (P = 0.002,

Mann-Whitney U test; Figure 1A), asymptomatic LRRK2 (median and IQR = 0.64 (0.43-0.8)

ng/ml, n = 50) (P < 0.0215, Mann-Whitney U test; Figure 1A), and symptomatic LRRK2 (median

and IQR = 0.61 (0.35-0.80) ng/ml, n = 21) (P < 0.0103, Mann-Whitney U test) patients (Figure

1A). There were no differences in the concentrations of CSF t-α-syn between the groups of sPD,

asymptomatic LRRK2 and symptomatic LRRK2 cases.

In contrast and as shown in Figure 1B, the levels of CSF o-α-syn were significantly higher

in the sPD group than in the HC group (median and IQR = 188 (171-220) pg/ml, n = 60) (P <

0.0001, Mann-Whitney U test; Figure 1B). Interestingly, CSF o-α-syn levels were also

significantly higher in the asymptomatic LRRK2 group than in the HC group (median and IQR =

182 (157-220) pg/ml, n = 50) (P < 0.006, Mann-Whitney U test; Figure 1B). Surprisingly, levels

of CSF o-α-syn in the symptomatic LRRK2 mutation carriers (median and IQR = 162 (145-183)

pg/ml, n = 21) were similar to those in the HC (median and IQR = 161 (148-186) pg/ml, n=39) (P

= 0.905, Mann-Whitney U test; Figure 1B), but significantly lower than CSF o-α-syn levels in

patients with sPD (P = 0.0008, Mann-Whitney U test) and in asymptomatic LRRK2 mutation

carriers (median and IQR = 182 (157-220) pg/ml, n=50).

Examination of CSF levels of pS129-α-syn revealed that CSF pS129 levels were higher in

patients with sPD and symptomatic LRRK2 compared to HC, but this was only significant for

sPD vs. HC (median and IQR = 139 (114-163) pg/ml, n=60) (P = 0.0281, Mann-Whitney U test;

Figure  1C).  

Table&2.b|&CSF&biomarkers&among&different&diagnostic&groups.&One&way&ANOVA&was&used&for&comparisions&among&all&thr&groups,&while&MannDWhitney&U&test&was&used&for&comparisons&between&the&two&groups&as&illustrated&in&the&table.

All#groups sPD#vs.#HC LRRK24HC#vs#HC LRRK24PD#vs.#HC LRRK24PD#vs.#sPDLRRK24PD#vs.#LRRK24HC

t4⍺4syn#(ng/ml) 0.0099 0.002 0.0215 0.0103 0.4982 0.3226o4⍺4syn#(pg/ml) 0.0001 <0.0001 0.006 0.9052 0.0008 0.0264pS1294⍺4syn#(pg/ml) 0.0835 0.0281 0.9338 0.5156 0.3414 0.5947o4/t4⍺4syn#% 0.0002 <0.0001 0.0012 0.0058 0.5365 0.9437pS1294/t4⍺4syn#% 0.0013 0.0003 0.0595 0.0019 0.862 0.1016

P&value

  137  

 

The ratio of o-α-syn to t-α-syn (o-/t-α-syn ratio) in the CSF was significantly higher in the

sPD (median and IQR = 33.1 (24.3-41.4) %, n = 60) (P < 0.0001, Mann-Whitney U test),

asymptomatic LRRK2 (median and IQR = 31.3 (21.6-45.9) %, n = 50) (P < 0.0012, Mann-Whitney

U test), and symptomatic LRRK2 (median and IQR = 28.7 (20.9-42.6) %, n = 21) (P < 0.0058,

Mann-Whitney U test) groups than in the age-matched HC group (median and IQR = 22.6 (15.4-

27.4) %, n = 39) (Figure 1D). The CSF pS129-/t-a-syn ratio was significantly higher in sPD

(median and IQR = 25.4 (16.4-30.2) %, n = 60) (P = 0.0003, Mann-Whitney U test), and in

symptomatic LRRK2 (median and IQR = 24.4 (18.1-29.7) %, n = 21) (P = 0.0019, Mann-Whitney

U test) groups than in the age-matched HC group (median and IQR = 15 (10.9-21.1) %, n = 39)

(Figure 1E), but was not significantly higher in asymptomatic LRRK2 (median and IQR = 20.3

(13-29.3) %, n = 50) (P = 0.0595, Mann-Whitney U test; Figure 1E).

 

Figure 1. Box and Whisker plots of CSF α-syn species levels among the diagnostic groups

   

   

*****

***

(A) *****

*****

(B)

***

(C)

****

****

(D)

  138  

 

CSF levels of total α-synuclein (ng/ml), (B)

CSF levels of oligomeric α-synuclein (pg/ml),

(C) CSF levels of pSer129-α-synuclein

(pg/ml), (D) CSF oligomeric/total α-synuclein

ratio %, (E) CSF phosphorylated S129/total α-

synuclein ratio %. The line through the middle

of the boxes corresponds to the median and the

lower and the upper lines to the 25th and 75th

percentile, respectively. The whiskers extend

from the 5th percentile on the bottom to the

95th percentile on top. Differences between

groups were assessed using Mann-Whitney U

–test, adjusted for age and gender. HC:

Healthy controls, sPD: sporadic PD, LRRK2-

HC: asymptomatic LRRK2 carriers, LRRK2-

PD: symptomatic LRRK2 carriers. * p<0.05,

** p<0.01, *** p<0.001

 

 

 

Correlations between CSF alpha-synuclein levels and clinical parameters

Correlations between CSF levels of α-syn species and the clinical profiles of the patients in both

sPD and symptomatic LRRK2 groups are summarized in Table 3. Higher levels of CSF t-α-syn

correlated with: worse cognitive function as assessed by the Montreal Cognitive Assessment

(MoCA) score (r= -0.48, P<0.001), longer disease duration (r= 0.26, P<0.05), in the sPD group,

and stronger impairment of motor functions as demonstrated by the UPDRS-III score (r= 0.52,

P<0. 01) in symptomatic LRRK2 carriers. However, there were no correlations between CSF o-

or pS129-α-syn levels and the clinical parameters we analyzed (Table 3).

***

**

(E)

  139  

Table 3| Spearman correlations between CSF alpha-synuclein species and clinical parameters

 

Subgroup analysis

PET image analysis

We assessed the correlation of PET data with the levels of CSF α-syn species in asymptomatic

LRRK2 mutation carriers (Table 4 and Figure 2). Based on their PET data, asymptomatic LRRK2

subjects were classified into four impairment stages by visual inspection: normal (n= 14), mild (n=

15), moderate (n= 14) and severe (n=7). Interestingly, by exploring the levels of the CSF α-syn

species among the different subgroups, we found that CSF t-α-syn levels were significantly higher

(P < 0.041, Kruskal-Wallis test; Figure 2A) when asymptomatic LRRK2 subjects were moderately

impaired, whereas subjects classified as severely impaired by PET scan analysis exhibited higher

levels of CSF o-α-syn (P < 0.048, Kruskal-Wallis test; Figure 2B). However, CSF pS129-α-syn

levels did not significantly change across the different stages (P = 0.438, Kruskal-Wallis test,

Figure 2C). The differences in the o-/t-α-syn ratio were more pronounced among the subgroups (P

= 0.02, Kruskal-Wallis test; Figure 2D) than the differences in the CSF pS129-/t-α-syn ratio (P =

0.678, Kruskal-Wallis test, Figure 2E).

Table&3|&Spearman&correlations&between&CSF&alpah7synuclein&species&and&clinical&parametersDisease&Duration H&Y UPDRS&III MoCA

Spordaic&PDt"⍺"syn&(ng/ml) 0.26* 0.14 0.16 "0.48***o"⍺"syn&(pg/ml) "0.04 "0.04 0.07 0.15pS129"⍺"syn&(pg/ml) 0.21 0.18 0.18 "0.10Symptomatic& LRRK2 &carrierst"⍺"syn -0.10 0.39 0.58** -0.42o"⍺"syn 0.32 0.35 0.33 "0.33pS129"⍺"syn 0.19 0.28 0.32 "0.15

  140  

Figure 2. Box and Whisker plots of CSF α-syn species in asymptomatic LRRK2 carriers

subgroups based on PET image analysis.

   

   

 

(A) CSF levels of total α-synuclein (ng/ml), (B) CSF levels of oligomeric α-synuclein (pg/ml), (C) CSF levels of pSer129-α-synuclein (pg/ml). The line through the middle of the boxes corresponds to the median and the lower and the upper lines to the 25th and 75th percentile, respectively. The whiskers extend from the 5th percentile on the bottom to the 95th percentile on top. Differences between groups were assessed using Kruskal-Wallis test, adjusted for age and gender. * p<0.05, ** p<0.01, *** p<0.001.  

 

(A) *

(B)

(C) (D) *

(E)

  141  

Table 4 | Biomarkers values in asymptomatic LRRK2 carriers’ subgroups (Median and

Interquartile range)

ROC analysis

ROC analysis was carried out for the CSF α-syn species, with each marker analyzed alone or in

combination with other markers to assess the power to discriminate sPD and LRRK2 groups from

the HC group (Figure 3). CSF o-α-syn achieved the best diagnostic performance as a single marker

to differentiate patients with sPD from HC (AUC= 0.74, sensitivity= 65%, specificity= 71%), with

CSF pS129-α-syn achieving the second highest diagnostic performance (AUC= 0.63, sensitivity=

61.7%, specificity= 71.5%). However, the o-/t-α-syn and pS129-/t-α-syn ratios performed even

better than the species alone, with AUCs of 0.76 and 0.71, sensitivities of up to 71.7% and 60%,

and specificities of up to 76.9% and 74.4%, respectively (see Figure 3A). The sensitivity and

specificity to discriminate symptomatic LRRK2 mutation carriers from HC were relatively poor

for most α-syn species alone (Figure 3C). However, the o-/t-α-syn and pS129-/t-α-syn ratios

achieved AUCs of 0.71 and 0.74, with sensitivities up to 61.9% and 71.4%, and specificities of up

to 71.4% and 66.7 respectively (Figure 3C). However, the sensitivity and specificity for CSF α-

syn species either alone or in combination to discriminate the asymptomatic LRRK2 group from

the HC group were relatively poor (Figure 3B).

Table&4&|&Biomarkers&values&in&asymptomatic&LRRK2&carriers&&subgroups&(Median&and&Interquatrile&range)Normal'(n=14) Mild'(n=15) Moderate'(n=14) Severe'(n=7) All'groups

t<⍺<syn'(ng/ml) 0.53'(0.29<'0.80) 0.58'(0.39<1.39) 0.76'(0.50<1.42)' 0.7'(0.28<'1.02) 0.0418o<⍺<syn'(pg/ml) 194'(155<'264) 178'(130<281) 160'(138<259) 192'(161<316) 0.0485pS129<⍺<syn'(pg/ml) 119'(62<'172) 110'(67<208) 135'(74<234) 120'(94<146) 0.4382o</t<⍺<syn'% 40.6'(19.3<91.7) 28.6'(13.0<61.6)' 22.8'(11.8<49.6) 41.9'(18.6<62.0) 0.0208pS129</t<⍺<syn'% 19.7'(7.9<53.6) 22.45'(5.7<48.9) 20.5'(6.2<40.9) 20'(12.0<36.3) 0.6782

  142  

Figure 3. Use of receiver operating characteristic (ROC) curves for the levels of CSF α-syn

levels in HC, sPD, LRRK2-HC and LRRK2-PD.

 

 

 

 

 

 

CSF t-α-syn, o-α-syn, pS129-α-syn, o-/t-α-syn ratio (%) and pS129-/t-α-syn ratio (%) to discriminate sPD patients from HC subjects (A), asymptomatic LRRK2 mutation carriers (LRRK2-HC) from HC subjects (B) and symptomatic LRRK2 mutation carriers (LRRK2-PD) from HC subjects (C). HC: Healthy controls, sPD: sporadic PD, LRRK2-HC: asymptomatic LRRK2 carriers, LRRK2-PD: symptomatic LRRK2 carriers. * p<0.05, ** p<0.01, *** p<0.001.

Descriminating,sPD,from,HCPredictor AUC Sensitivity Specificity P value

T-α-syn (ng/ml) 0.318 51.7 23.1 0.002

O-α-syn (pg/ml) 0.741 65.0 71.8 0.000

pS129-α-syn (pg/ml) 0.631 61.7 61.5 0.028

O-/T-α-syn % 0.762 71.7 76.9 0.000

pS129-/T-α-syn % 0.714 60.0 74.4 0.000

Descriminating,LRRK21HC,from,HCPredictor AUC Sensitivity Specificity P value

T-α-syn (ng/ml) 0.358 41.2 43.6 0.022

O-α-syn (pg/ml) 0.668 68.6 56.4 0.006

pS129-α-syn (pg/ml) 0.505 58.8 46.2 0.932

O-/T-α-syn % 0.697 60.8 69.2 0.001

pS129-/T-α-syn % 0.616 60.8 56.4 0.059

Descriminating,LRRK21PD,from,HCPredictor AUC Sensitivity Specificity P valueT-α-syn (ng/ml) 0.300 42.9 35.9 0.011

O-α-syn (pg/ml) 0.490 52.4 51.3 0.901

pS129-α-syn (pg/ml) 0.552 57.1 48.7 0.510

O-/T-α-syn % 0.715 61.9 74.4 0.006

pS129-/T-α-syn % 0.741 71.4 66.7 0.002

(A)

(B)

(C)

  143  

To further explore how cognitive functions can contribute to the segregation of PD patients from

HC subjects, our sPD cohort were classified into 3 subgroups based on the MoCA score: (1) sPD

not cognitively impaired (n = 34, MoCA >26), (2) mild cognitive impairment (MCI) sPD (n = 21,

MoCA 22-26), and (3) sPD with dementia (PDD) (n = 5, MoCA <22). Remarkably, assessing the

ROC curves for the o-/t-α-syn and pS129-/t-α-syn ratios for patients with PD without any cognitive

impairment generated better rates of sensitivity and specificity than were found for the overall

mixed sPD group (AUC= 0.84, 0.78; sensitivity= 83.3%, 73.3%; and specificity= 82.1%, 74.4%;

respectively) (Supplementary Figure 1). However, the ROC curves generated for the MCI and

PDD subgroups showed poor diagnostic performance in terms of the sensitivity and specificity

(Figure 4).

Figure  4.  Use  of  receiver  operating  characteristic  (ROC)  curves  for  the  levels  of  CSF  α-­‐syn  

levels    in  sPD  sub-­‐groups.  

 

 

 

 

Descriminating,Pure,PD,from,HCPredictor AUC Sensitivity Specificity P value

T-α-syn (ng/ml) 0.229 23.3 41.0 0.000

O-α-syn (pg/ml) 0.761 76.7 59.0 0.000

pS129-α-syn (pg/ml) 0.614 70.0 53.8 0.107

O-/T-α-syn % 0.842 83.3 82.1 0.000

pS129-/T-α-syn % 0.775 73.3 74.4 0.000

Descriminating,MCI,PD,from,HCPredictor AUC Sensitivity Specificity P valueT-α-syn (ng/ml) 0.403 42.9 43.6 0.218

O-α-syn (pg/ml) 0.724 57.1 71.8 0.004

pS129-α-syn (pg/ml) 0.663 61.9 69.2 0.039

O-/T-α-syn % 0.676 57.1 69.2 0.025

pS129-/T-α-syn % 0.660 52.4 79.5 0.042

(A)

(B)

  144  

CSF t-α-syn, o-α-syn, pS129-α-syn, o-/t-α-syn ratio (%) and pS129-/t-α-syn ratio (%) to

discriminate pure sPD patients from HC subjects (A), MCI sPD from HC (B) and demented sPD

from HC subjects (C). * p<0.05, ** p<0.01, *** p<0.001

 

Discussion

Several reasons underline why asymptomatic LRRK2 mutation carriers are ideal candidates for

the exploration of surrogate biomarkers for early detection of PD (1) the known long duration of

the prodromal or premotor phase, (2) the early dopaminergic dysfunction demonstrated by

abnormal PET changes and (3) once converted, the clinical phenotype is similar to sporadic PD.

The aim of this study was therefore to explore the potential of CSF α-syn species as biomarkers

for early detection of PD, preferably at the preclinical stage in asymptomatic LRRK2 mutation

carriers as in comparison to sPD cases.

The reduction in CSF t-α-syn and the increase of CSF o- and pS129-α-syn levels in the

sPD group relative to the controls is in agreement with findings in previous independent reports.3,

12 In a recent study, Vilas et al.13 found a significant decrease in CSF t-α-syn levels in sPD patients

compared to symptomatic LRRK2 mutation carriers, whereas, in the current study we observed

significant decrease in the levels of CSF t-α-syn in symptomatic LRRK2 cases compared to HC.

In spite of the small number of LRRK2 carriers, a significant increase was found in the CSF o-α-

syn levels in asymptomatic LRRK2 mutation carriers compared to those in HC subjects, an increase

that was apparent but not significant in our previous study.5 This discrepancy most likely can be

attributed to the use of a new highly specific oligomeric ELISA in the present study, which

demonstrated its sensitivity and robustness compared to the first-generation assay.3 The  significant  

elevation of CSF o-α-syn in asymptomatic LRRK2 subjects compared to HC supports the

hypothesis that CSF o-α-syn is a potential biomarker for the early detection of PD.

Multiple lines of evidence suggest that oligomerization is a very early event in PD

pathogenesis and that o-α-syn is also thought to be the precursors of formed LBs, and the culprits

for neuronal degeneration in PD. While at the later stages of PD α-syn oligomers are further

maturated into fibrillar form and sequestered away into LBs. This could explain the high levels of

CSF o-α-syn detected in asymptomatic LRRK2 subjects, whereas, low levels of CSF o-α-syn in

LRRK2 patients, considering the long disease duration and the severe motor-activity decline noted

in the latest group.

  145  

The value of using a panel of different markers rather than a single marker has become

clearer in the clinical practice and is well important also due to the clinical heterogeneity of PD.

When we combined the analyses of CSF t- and o-α-syn and measured the ratio of these species,

the AUC value representing the ability to discriminate between patients with sPD and HC was

0.74 for CSF o-α-syn alone but improved to 0.76 for the o-/t-α-syn ratio. Similarly, the AUC

increased from 0.63 for CSF pS129-α-syn alone to 0.71 for the ratio. However, the o-/t-α-syn and

pS129-/t-α-syn ratios exhibited AUCs of 0.71 and 0.74, respectively, to discriminate asymptomatic

LRRK2 mutation carriers from HC.

Several cross-sectional and longitudinal cohort studies correlated increased levels of CSF

t-α-syn with cognitive decline or dementia3, 14 but not with motor impairment in PD. Elevated

levels of CSF t-α-syn have been reported in prion disease and Alzheimer’s disease patients

compared to those in HC, and t-α-syn levels have been positively correlated with tau species and

negatively correlated with the Mini-Mental State Examination (MMSE) score in Alzheimer’s

disease.15-17 Therefore, we sub-grouped sPD patients based on their MoCA scores and assessed

the power of CSF α-syn forms in segregating pure PD patients with no signs of cognitive decline

from HC group. As expected, MCI and PDD patients corresponded to higher CSF t-α-syn levels

compared to pure PD patients. Interestingly, CSF o-/t-α-syn ratio displayed better diagnostic

performance at discriminating pure PD patients rather than the mixed sPD population from HC

subjects. This observation highlights the significant role of CSF o-/t-α-syn ratio in stratifying PD

patients for biomarkers discovery and perhaps for clinical trials that specifically target α-syn.  

Future studies in large-scale, prospective and well-controlled studies, where presymptomatic

LRRK2 subjects are followed up longitudinally up to the appearance of the clinical symptoms such

as the ongoing PPMI will further validate the findings reported in our current study.

In summary, the assessment of the diagnostic performance of CSF α-syn species across

different study groups, CSF o-α-syn were significantly elevated in asymptomatic LRRK2 subjects

compared to symptomatic LRRK2 patients. CSF o-/t-α-syn ratio attained better sensitivity and

specificity scores rather than single species, further supporting the optimum diagnostic utility of

CSF biomarkers ratios. Although weak, but present, CSF o-α-syn biomarkers correlated with PET

scan images in asymptomatic LRRK2 mutation carriers. Importantly, our findings suggest that

future biomarkers discovery studies should consider stratifying the selected PD cohort by

separating patients with cognitive impairment and possibly other phenotypes, that have to be

  146  

identified. To account for the different phenotype and the vast heterogeneity of PD also other

marker candidates need to be identified that can add to the synuclein marker presented here. Future

studies will further validate our findings in larger cohorts, where asymptomatic LRRK2 subjects

and early diagnosed sPD patients are followed up longitudinally to detect the early appearance of

motor symptoms and track the disease progression.

Acknowledgments

The authors thank all patients and healthy subjects, lab manager Dr. Houari Abdesselem, and

funding agencies. This work would have not been possible without their valuable contribution.

Author contributions

Study design and supervision: N.M, O.E. Conduction of the experiments: N.M, M.T., M.F. and

N.V. Clinical data collection: J.A. and E.H. Data analysis: N.E. and N.M. Writing the first draft of

the manuscript and incorporating revisions from other authors: N.M. Data interpretation and

critical revision of the manuscript for important intellectual content: N.M, J.A., W.B., T.T., B.M.,

H.B., and O.E. Final review of the draft and approval to submit for publication: O.E. All authors

read and approved the final manuscript.

 

   

  147  

References

1.   Wu  YR,  Chang  KH,  Chang  WT,  et  al.  Genetic  variants  ofLRRK2  in  Taiwanese  Parkinson's  disease.  PloS  one  2013;8:e82001.  2.   Stewart   T,   Sossi   V,   Aasly   JO,   et   al.   Phosphorylated   α-­‐synuclein   in   Parkinson's   disease:  correlation  depends  on  disease  severity.  Acta  Neuropathol  Commun  2015;3:7.  3.   Majbour   NK,   Vaikath   NN,   van   Dijk   KD,   et   al.   Oligomeric   and   phosphorylated   alpha-­‐synuclein   as   potential   CSF   biomarkers   for   Parkinson's   disease.  Molecular   neurodegeneration  2016;11:7.  4.   Aasly  JO,  Toft  M,  Fernandez-­‐Mata  I,  et  al.  Clinical  features  of  LRRK2-­‐associated  Parkinson's  disease  in  central  Norway.  Annals  of  neurology  2005;57:762-­‐765.  5.   Aasly  JO,  Johansen  KK,  Brønstad  G,  et  al.  Elevated  levels  of  cerebrospinal  fluid  α-­‐synuclein  oligomers  in  healthy  asymptomatic  LRRK2  mutation  carriers.  Front  Aging  Neurosci  2014;6:248-­‐248.  6.   Aasly   JO,   Vilarino-­‐Guell   C,   Dachsel   JC,   et   al.   Novel   pathogenic   LRRK2   p.Asn1437His  substitution   in   familial   Parkinson's   disease.   Movement   disorders   :   official   journal   of   the  Movement  Disorder  Society  2010;25:2156-­‐2163.  7.   Johansen  KK,  Hasselberg  K,  White  LR,  Farrer  MJ,  Aasly  JO.  Genealogical  studies  in  LRRK2-­‐associated  Parkinson's  disease  in  central  Norway.  Parkinsonism  &  related  disorders  2010;16:527-­‐530.  8.   Johansen  KK,  White  LR,  Farrer  MJ,  Aasly  JO.  Subclinical  signs  in  LRRK2  mutation  carriers.  Parkinsonism  &  related  disorders  2011;17:528-­‐532.  9.   Sossi  V,  de  la  Fuente-­‐Fernandez  R,  Nandhagopal  R,  et  al.  Dopamine  turnover  increases  in  asymptomatic  LRRK2  mutations  carriers.  Movement  disorders  :  official  journal  of  the  Movement  Disorder  Society  2010;25:2717-­‐2723.  10.   Vaikath  NN,  Majbour  NK,  Paleologou  KE,  et  al.  Generation  and  characterization  of  novel  conformation-­‐specific   monoclonal   antibodies   for   alpha-­‐synuclein   pathology.   Neurobiology   of  disease  2015;79:81-­‐99.  11.   Majbour  NK,   Vaikath  NN,   Eusebi   P,   et   al.   Longitudinal   changes   in   CSF   alpha-­‐synuclein  species   reflect   Parkinson's   disease   progression.   Movement   disorders   :   official   journal   of   the  Movement  Disorder  Society  2016;31:1535-­‐1542.  12.   Park  MJ,  Cheon  S-­‐M,  Bae  H-­‐R,  Kim  S-­‐H,  Kim  JW.  Elevated  levels  of  α-­‐synuclein  oligomer  in  the  cerebrospinal  fluid  of  drug-­‐naïve  patients  with  Parkinson's  disease.  J  Clin  Neurol  2011;7:215-­‐222.  13.   Vilas  et  al.  14.   Stewart   T,   Liu   C,   Ginghina   C,   et   al.   Cerebrospinal   fluid   α-­‐synuclein   predicts   cognitive  decline  in  Parkinson  disease  progression  in  the  DATATOP  cohort.  Am  J  Pathol  2014;184:966-­‐975.  15.   Majbour   NK,   Chiasserini   D,   Vaikath   NN,   et   al.   Increased   levels   of   CSF   total   but   not  oligomeric   or   phosphorylated   forms   of   alpha-­‐synuclein   in   patients   diagnosed   with   probable  Alzheimer's  disease.  Sci  Rep  2017;7:40263-­‐40263.  16.   Kasai  T,  Tokuda  T,  Ishii  R,  et  al.  Increased  alpha-­‐synuclein  levels  in  the  cerebrospinal  fluid  of  patients  with  Creutzfeldt-­‐Jakob  disease.  Journal  of  neurology  2014;261:1203-­‐1209.  

  148  

17.   Llorens  F,  Kruse  N,  Schmitz  M,  et  al.  Evaluation  of  alpha-­‐synuclein  as  a  novel  cerebrospinal  fluid  biomarker  in  different  forms  of  prion  diseases.  Alzheimer's  &  dementia  :  the  journal  of  the  Alzheimer's  Association  2017;13:710-­‐719.    

   

  149    

!!

CHAPTER 7

SUMMARY & GENERAL DISCUSSION

  150  

CHAPTER 7 | SUMMARY & GENERAL DISCUSSION  Biomarkers for Parkinson’s disease (PD) are of paramount importance, since the clinical symptoms are

often nonspecific and show overlap with other neurodegenerative diseases, the clinical progression is highly

variable, and multiple clinical phenotypes exist for the same disease [1]. Primary endpoints of most clinical

trials in PD depend on measures of motor and cognitive impairment, which are critically hindered by a lack

of disease specificity or sensitivity [2]. Biomarkers reflecting the underlying disease mechanisms at the

molecular level might prove useful in improving diagnostic accuracy, monitoring disease progression, and

serving as surrogate endpoints in clinical trials. However, at this point we are still far beyond having solid

biomarkers for PD.

In this thesis, we address the challenges for the identification of informative body fluid biomarkers for PD,

and we highlight the effort made to develop robust immunoassays targeting different species of alpha-

synuclein (α-syn), aiming to provide a panel of disease-reflecting biomarkers for the early diagnosis of PD,

and for the monitoring of disease progression. The recognition that α-syn protein is abundant in Lewy

bodies (LBs) [3], the finding that mutations in the gene encoding α-syn protein (SNCA) [4], as well as that

duplication and triplication of this gene lead to early-onset, familial PD [5], and the reports of transgenic

flies and animals with human wild-type or mutant α-synuclein genes developing neuronal dysfunction and

PD-like pathology [6] were behind the growing interest in α-syn as a critical player in PD pathogenesis.

After a 20-year journey of α-syn research [7], we have come to understand that α-syn can adopt several

different pathological conformations. In addition, α-syn can be detected in extracellular fluids, such as CSF,

blood and saliva [8, 9]. These findings prompted us to explore the role of α-syn as a diagnostic and

progression biomarker for PD.

In this chapter, we recount and discuss our discoveries from the previous chapters of this thesis. Thereafter,

we summarize our conclusions concerning the potential role of α-syn species as diagnostic and progression

biomarkers for PD. We then describe how our findings can facilitate reaching the goal of an early diagnosis

of PD and pave the way towards the development of more effective therapeutic strategies for the disease.

At the end of the chapter, we comment on future directions for biomarker discovery in PD and related

disorders.

In Chapter 1, we reviewed the progress made to date towards the development and validation of body fluid

biomarkers to improve the diagnosis and treatment of PD. Despite the progress made in the past decades in

understanding the etiology of PD [10], the diagnosis still depends on clinical criteria, hindering the early

detection of a disease that has a long prodromal or premotor phase. The existing literature was reviewed

looking for the answer to the most important question; What might be the ideal biomarker for PD? While

the answer is yet to be found, there are reasons to be believe that we are getting closer to that goal. Perhaps,

  151  

the most insightful conclusions made in Chapter 1, are: 1) a panel of biomarkers rather than a single marker

is the way forward, 2) some biomarkers better differentiate PD from other forms of parkinsonism (NF-L

levels), while others better predict PD progression (CSF t-a-syn, CSF o-a-syn), 3) it is important to adhere

to strictly and thoroughly standardized protocols for body fluid sample collection and processing to reach

consistent results.

Considering the minute levels of the different species of α-syn in body fluids, studying their presence in

different biological compartments of the human body in association with the pathophysiology of PD

demands robust tools with high specificity and affinity. The optimal way to address these challenges was

through the generation of conformation-specific monoclonal antibodies (mAbs) that can specifically target

PD pathology. This aim was successfully achieved in Chapter 2, where we described the generation and

characterization of conformation-specific mAbs against α-syn aggregates. Our mAbs were generated

following the hybridoma technology, where recombinant α-syn aggregates were used to immunize female

balb/C mice. Only IgG isotype clones that showed specificity towards α-syn aggregates were selected for

further characterization. The specificity of the mAbs towards α-syn aggregates was assessed by means of

inhibition ELISA, dot blotting, and preadsorption experiments. Our mAbs did not cross-react with

monomeric or fibrillar forms of β- or γ-syn, or any other amyloidogenic proteins or peptides such as Aβ,

Tau40, IAPP or ABri. This clearly established that although our mAbs were raised against a conformational

epitope, they are still highly selective, unlike many other conformation-specific mAbs that recognize the

conformation of multiple amyloid proteins [11]. To ensure that the affinity of our mAbs towards α-syn

aggregates would outreach their peer antibodies, surface plasmon resonance analysis was deployed where

most of our antibodies showed affinity towards α-syn fibrils in the picomolar range. In an attempt to acquire

more details about the epitope of our mAbs, pepscan using a peptide library spanning the entire α-syn

sequence was performed. MAbs exhibited very weak reactivity for the C-terminal peptide spanning the α-

syn sequence between amino acids 127 and 140, confirming that our mAbs do not recognize linear peptides

and that the C-terminus of α-syn may be a part of the complex conformational epitope. The potential of the

mAbs was additionally explored by staining human brain sections of patients with PD, dementia with Lewy

bodies (DLB), Alzheimer’s disease (AD), and multiple system atrophy (MSA) along with healthy control

cases. Immunohistochemical analysis showed that our mAbs were able to recognize α-syn pathology in

synucleinopathies, as well as identify pathological structures that were not detected by other commercially

available antibodies. In summary, Chapter 2 detailed the generation and characterization of six novel

conformation-specific mAbs against PD pathology. These mAbs can either be exploited to develop

immunoassays that may serve as diagnostic tools for PD, or be engineered to serve as therapeutic agents in

the form of vaccines in the field of passive immunotherapy [12].

  152  

Our quest for developing robust tools that can improve our understanding of PD pathology did not stop

there. In Chapter 3, we went on to use the mAbs described in Chapter 2 for the development of

quantitative analytical methods, which resulted in highly sensitive and specific enzyme-linked

immunosorbent assays (ELISA). In this chapter, we additionally reported the characterization of more

mAbs (11D12, and PS129), as well as our polyclonal antibody Syn-140. We also elaborated on the

sensitivity and specificity of the three ELISA assays that were developed to capture total- (t-), oligomeric-

(o-), or phosphorylated S129 (p-S129-) a-syn. Chapter 3 highlights the value of adding a biochemical test

to the diagnostic procedures in PD to overcome the limitations of current clinical criteria and reduce the

high rates of misdiagnosis in the early stages of the disease. We explored the potential of our novel assays

for the discovery of CSF biomarkers in a Dutch cohort of PD patients and age matched healthy controls.

Analyses of this cross-sectional cohort revealed reduced CSF t-α-syn, and elevated CSF o- and p-S129-α-

syn levels in PD compared to healthy controls. Highlighting the diagnostic power of multiple CSF

biomarkers, combining both o-/t-α-syn and p-S129-/t- α-syn ratios improved the ability to discriminate

between PD and healthy controls, which was further enhanced by including p-tau. When investigating the

correlations of biomarker levels with disease severity in the PD group, CSF t-α-syn levels inversely

correlated with MMSE scores, whereas CSF o-α-syn levels inversely correlated with H&Y scores. In

conclusion, the results described in Chapter 3, 1) highlight the potential of our biomarker assays in the

field of PD and other synucleinopathies, 2) address the shortcomings of previous ELISA platforms [13](that

used a generic antibody for capture, and the biotinylated form for detection) by using a novel conformation-

specific mAb (Syn-O2), and 3) validate the usefulness of combining multiple CSF biomarkers in improving

diagnostic accuracy in PD.

To take full advantage of the merits of the assays described in Chapters 2 and 3, and to expand our

understanding of the role that α-syn species may have as progression biomarkers in PD, we measured the

different forms of α-syn (t, o- and p-S129-α-syn) in CSF from 121 participants in the longitudinal Deprenyl

and Tocopherol Antioxidative Therapy for Parkinsonism (DATATOP) study cohort, who were clinically

diagnosed with early-stage PD (probability between 90%-100%), which is described in Chapter 4.

DATATOP is a multicenter, placebo-controlled clinical trial involving patients with early-stage PD, who

underwent repeated CSF sampling during a 2-year period, prior to starting dopamine replacement therapy

[14]. We examined the changes in CSF t-, o-, p-S129-α-syn, t-tau, p-tau, Aβ 1-40, and Aβ 1-42 during the

2-year follow-up period. Compared to baseline levels, follow-up CSF levels of t- and o-α-syn levels were

significantly higher, whereas p-S129-α-syn levels were significantly lower. In addition, the changes in t-

and o-α-syn levels, but not p-S129-α-syn levels positively correlated with duration of follow-up, confirming

the increase in CSF t- and o-α-syn levels with disease progression. Although changes in CSF t- or o-α-syn

levels alone did not correlate with clinical measures of motor progression, the change in the ratio of o-/t-α-

  153  

syn was associated with worsening UPDRS motor scores. Interestingly, patients with predominant postural

instability and gait difficulty revealed a stronger correlation between the change in the o-/t-α-syn ratio and

the change in UPDRS motor scores compared to other clinical phenotypes, defined as tremor dominant or

intermediate subtypes, thus coupling the CSF biochemical profile of α-syn species with the clinical

phenotype of PD. The data in Chapter 4 shed light on the dynamic pattern of change in CSF α-syn species

over the course of the disease, supporting their potential role as disease progression biomarkers and their

association with PD clinical phenotypes. The findings in Chapters 3 and 4 together demonstrate the

potential role of α-syn species as surrogate biomarkers for PD diagnosis and disease progression

monitoring. Despite the limitations surrounding the DATATOP cohort,   including long storage of CSF

samples (> 25 years), no tandardized protocols for sample collection across participating centers, and the

lack of a control group, our findings are of paramount importance especially in the context of PD

progression. Clearly, to fully appreciate the role CSF α-syn species can play in monitoring PD progression,

our findings should be extended to larger cohorts where PD patients are followed longitudinally until

autopsy, providing a definitive neuropathological confirmation of the clinical diagnosis.

The co-existence of α-syn and tau pathology observed in the brains of patients with AD, PD and DLB[15,

16], prompted us to study the association between CSF α-syn and tau in an AD cohort. The results of this

study, described in Chapter 5, demonstrated an improvement of diagnostic sensitivity/specificity of

classical AD CSF biomarkers when adding CSF α-syn species. Using the same immunoassays described in

Chapters 3 and 4, we measured CSF levels of different α-syn species in an Italian cohort of AD patients,

who showed a CSF profile typical of AD at baseline, as well as in cognitively intact controls. The AD

patients in this cohort were selected based on both clinical evaluation and a CSF profile of Aβ-42, t-tau and

p-tau supporting the clinical diagnosis of AD. CSF t-α-syn levels were significantly higher in the AD group

compared to the control group, whereas CSF o- and p-S129-α-syn levels were not significantly different

between the groups. A positive correlation between t-α-syn and tau species was found only in the AD group.

The role of t-α-syn in AD pathology taken together with the lack of o- and p-S129-α-syn contribution to

AD diagnosis, further support the hypothesis that o- and p-S129-α-syn species are intimately connected to

the pathogenesis of synucleinopathies rather than tauopathies. To broaden our understanding of the

potential role of α-syn as a cognitive biomarker [17], cohorts of subjects with α-syn related dementias,

including DLB and PD dementia, should be investigated [18]. Furthermore, combining α-syn and AD core

biomarkers with brain imaging data might contribute to the identification of individuals at higher risk of

MCI and conversion to AD. In conclusion, the results in Chapter 5 obtained in a well-characterized cohort

of AD patients and neurological controls further support the notion that t-α-syn most likely represents a

marker of neuronal loss and/or synaptic failure in AD [19].

  154  

While most cases of PD occur in a sporadic manner, a subset of cases are attributable to genetic causes.

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common causes of inherited PD.

In Chapter 6, we aimed to determine whether any of the CSF α-syn species have the potential to serve as

preclinical biomarkers for PD. Using our in-house developed ELISA assays, we measured t-, o- and pS129-

α-syn species in a well-characterized Norwegian cohort of 72 subjects with LRRK2 mutations: 21

symptomatic cases already diagnosed with PD and 51 asymptomatic LRRK2 mutation carriers. In parallel,

we also included 60 patients with sporadic PD (sPD) and 31 healthy control subjects. The observations in

this cohort suggested that CSF o-α-syn holds potential as a surrogate biomarker for the early detection of

PD. In addition, we observed a correlation between body fluid biomarker levels and PET scan images in

asymptomatic LRRK2 mutation carriers. Larger cohorts, where asymptomatic LRRK2 subjects are followed

up longitudinally, would further validate these findings.

Interpreting the findings from Chapters 3-5 together, the reduction of CSF t-α-syn in PD is most likely due

to the protein folding tendency and its subsequent sequestration in LBs, while the increased CSF o-α-syn

levels are most likely related to clearance failure of the oligomerized forms of α-syn. The scarce longitudinal

data challenge our speculations about the reasons behind the longitudinal changes in CSF t- and o-α-syn

levels over the course of PD. Elevated levels of α-syn have been linked to degree of neurodegeneration and

a worse prognosis of PD [20]. Having said that, only a weak inverse correlation between CSF o-α-syn and

motor impairment, and a moderate inverse correlation between t-α-syn and cognitive function were noted

in Chapter 3, while the levels of neither of these CSF α-syn species correlated with disease duration.

Longitudinal follow-up of cases, as described in Chapter 4, did show a correlation between the changes in

t- and o-α-syn levels and disease duration, a correlation that may grow stronger with a longer follow-up

period. In interpreting our findings, we have to keep in mind that PD is a disease that develops slowly over

many years. The average follow-up duration of two years in most longitudinal studies may be too short to

capture the true strength of the relationship between body fluid biomarkers and disease progression in PD.

As discussed in the previous chapters of this thesis, α-syn is subject to several post-translational

modifications (PTMs) [21], where phosphorylation at Serine 129 is among the most frequent ones [22]. So

far, we do not know exactly in what way α-syn phosphorylation alters the folding propensity or

neurotoxicity of the protein [23]. The fact that α-syn in LBs is predominantly phosphorylated at Ser129

[24] encouraged us to think that developing immunoassays measuring CSF p-S129-α-syn would best reflect

LB pathology. This hypothesis was supported by findings in cross-sectional studies by our group and others,

where higher levels of CSF p-S129-α-syn were found in PD [25, 26]. However, longitudinal studies have

come up with conflicting results. Stewart et al. reported a longitudinal increase in CSF p-S129-α-syn levels,

while we found a longitudinal decrease in cases from the DATATOP study [26]. Although differences in

  155  

selection criteria of the patients included in each study may explain these differences, this sparked a

controversy about the potential role of p-S129-α-syn as a PD biomarker.

In AD, the increase in CSF t-α-syn can be attributed to protein over-expression and/or excessive neuronal

damage in AD patients [27, 28], leading in turn to α-syn leakage from damaged cells into the brain’s

interstitial fluid and then into the CSF. This leakage, however, was not reflected in the levels of other forms

of α-syn, such as o- or p-S129-α-syn.

Among the different species of α-syn explored in this thesis, and the original findings these cohorts yielded,

the data suggest that the link between CSF α-syn biomarkers and PD severity can be summarized as follows:

1) CSF o-α-syn rather than t- or p-S129-α-syn is more connected to PD motor severity, 2) CSF t-α-syn is

associated with cognitive decline in PD, 3) CSF o-α-syn holds the biggest potential for early detection of

PD.  Large-scale, prospective studies in well-controlled cohorts, where CSF samples are collected and

processed using rigorously standardized procedures are needed to further validate these findings [29].

As highlighted in Chapter 1, several studies have reported inconsistent findings on α-syn levels among the

different labs suggesting there are gaps in our knowledge that should be filled. This has led research groups

to collaborate in order to identify the best strategies to move forward with body fluid biomarker studies.

This includes an increasing number of recent studies directed towards understanding the leading cause for

the lack of reproducibility between laboratories [30]. Many factors contributing to this inconsistency have

already been identified, such as: 1) protocols for biofluid collection and processing (storage duration, types

of tubes, centrifugation, etc.), 2) CSF contamination with blood traces, and hemoglobin levels in

serum/plasma, 3) immunoassays platforms, and 4) patient selection criteria.

Future Directions There is an increasing body of knowledge suggesting that multiple forms of α-syn can improve the

diagnostic accuracy of PD, especially when used as a panel of biomarkers and supported by neuroimaging

and clinical data. However, further studies using validated quantitative platforms and including patients

with different synucleinopathies that are followed longitudinally until autopsy are highly needed. When

further deployed in larger cohorts with longer follow-up durations, the assays we developed can facilitate

the search for reliable CSF biomarkers that correlate with disease severity. We will make our assays widely

available for the research community to help provide us with objective data from different biomarkers

discovery studies to assist in making informed decisions about the diagnosis in individual patients in a

timely manner. An accurate early diagnosis is desperately needed in the development of PD-specific

therapies to be able to distinguish PD cases from patients suffering from other synucleinopathies or forms

of parkinsonism that require different therapies.

  156  

Although considerable research is needed to translate α-syn biomarkers from the research setting to

diagnostic strategies, it is worth acknowledging the rapid advances that have been made in the field of PD

biomarkers, and the work towards accelerating the pace of developing disease modifying therapies for PD

and related disorders. Given the association of multiple proteins and markers other than α-syn with PD,

such as tau, Aβ42, neurofilament light chain, or inflammatory markers, it is very likely that the search for

an ideal panel of biomarkers will continue. Furthermore, multiple large-scale, longitudinal studies in well-

controlled cohorts, where standardized protocols are followed, like the Parkinson's Progression Markers

Initiative, are now underway and will yield important new data within the next decade. In addition, an

increasing number of studies have extended their search for biomarkers far beyond CSF and blood to also

include peripheral tissues such as submandibular salivary glands [31] and skin biopsies [32]. Although this

approach has not yet culminated in complete success, it might facilitate reaching the ultimate goal of an

ideal (panel of) biomarker(s) for PD in the near future for early and differential diagnosis, and monitoring

disease progression in PD.

In summary, while our understanding of PD over the past two centuries has vastly progressed and many

potential biomarkers have come to the fore, the ideal biomarker or panel of biomarkers for PD has yet to

be validated. On the bright side, multiple large-scale, longitudinal studies in well-controlled cohorts, where

standardized protocols are followed, like the Parkinson's Progression Markers Initiative, are now underway

and will yield important new data within the next decade.

Key findings of the thesis 1. Conformation-specific mAbs for α-syn aggregates were generated and employed to identify α-syn

pathology compared to pan antibodies;

2. 11D12 and Syn-140 antibodies were generated and served as good candidates for t-α-syn detection,

whereas PS129 mAb was specific for p-S129-α-syn;

3. Robust, sensitive and specific ELISA assays for t-, o-, or p-Ser129-α-syn forms were developed

and characterized;

4. In PD patients, CSF t-α-syn levels were significantly decreased, whereas CSF o-α-syn and p-s129-

α-syn levels were significantly increased compared to healthy controls;

5. The combination of CSF o-/t-α-syn, p-S129-α-syn and p-tau provided the best fitting predictive

model for discriminating PD patients from healthy controls;

6. CSF o-α-syn levels correlated with the severity of PD motor symptoms;

7. CSF t- and o- α-syn levels significantly increased over a two-year follow-up period in early-stage

PD, and the CSF o-/t-α-syn ratio was associated with the change in UPDRS motor scores;

  157  

8. CSF t-α-syn levels were significantly higher in AD patients compared to non-demented controls,

and corresponded to worsening of cognitive functions in the AD patients;

9. Levels of CSF t-α-syn were comparable among sporadic PD, symptomatic and asymptomatic

LRRK2 mutation carriers, but significantly higher than in healthy subjects;

10. CSF o-α-syn levels were significantly higher in asymptomatic LRRK2 subjects than in healthy

controls or symptomatic LRRK2 patients;

  158  

References

[1]   L.  Parnetti  et  al.,  "Cerebrospinal  fluid  biomarkers  in  Parkinson  disease,"  (in  eng),  Nat  Rev  Neurol,  vol.  9,  no.  3,  pp.  131-­‐40,  Mar  2013.  

[2]   L.  Marsili,  G.  Rizzo,  and  C.  Colosimo,  "Diagnostic  Criteria  for  Parkinson's  Disease:  From  James  Parkinson  to  the  Concept  of  Prodromal  Disease,"  (in  eng),  Front  Neurol,  vol.  9,  p.  156,  2018.  

[3]   M.  G.  Spillantini,  M.  L.  Schmidt,  V.  M.  Lee,  J.  Q.  Trojanowski,  R.  Jakes,  and  M.  Goedert,  "Alpha-­‐synuclein  in  Lewy  bodies,"  (in  eng),  Nature,  vol.  388,  no.  6645,  pp.  839-­‐40,  Aug  28  1997.  

[4]   P.  Ibáñez  et  al.,  "Causal  relation  between  alpha-­‐synuclein  gene  duplication  and  familial  Parkinson's  disease,"  (in  eng),  Lancet,  vol.  364,  no.  9440,  pp.  1169-­‐71,  2004  Sep  25-­‐Oct  1  2004.  

[5]   M.  C.  Chartier-­‐Harlin  et  al.,  "Alpha-­‐synuclein  locus  duplication  as  a  cause  of  familial  Parkinson's  disease,"  (in  eng),  Lancet,  vol.  364,  no.  9440,  pp.  1167-­‐9,  2004  Sep  25-­‐Oct  1  2004.  

[6]   H.  Mizuno,  N.  Fujikake,  K.  Wada,  and  Y.  Nagai,  "α-­‐Synuclein  Transgenic  Drosophila  As  a  Model  of  Parkinson's  Disease  and  Related  Synucleinopathies,"  (in  eng),  Parkinsons  Dis,  vol.  2011,  p.  212706,  2010.  

[7]   M.  Goedert,  R.  Jakes,  and  M.  G.  Spillantini,  "The  Synucleinopathies:  Twenty  Years  On,"  (in  eng),  J  Parkinsons  Dis,  vol.  7,  no.  s1,  pp.  S53-­‐S71,  2017.  

[8]   O.  M.  El-­‐Agnaf  et  al.,  "Alpha-­‐synuclein  implicated  in  Parkinson's  disease  is  present  in  extracellular  biological  fluids,  including  human  plasma,"  (in  eng),  FASEB  J,  vol.  17,  no.  13,  pp.  1945-­‐7,  Oct  2003.  

[9]   I.  Devic  et  al.,  "Salivary  α-­‐synuclein  and  DJ-­‐1:  potential  biomarkers  for  Parkinson's  disease,"  (in  eng),  Brain,  vol.  134,  no.  Pt  7,  p.  e178,  Jul  2011.  

[10]   S.  Przedborski,  "The  two-­‐century  journey  of  Parkinson  disease  research,"  (in  eng),  Nat  Rev  Neurosci,  vol.  18,  no.  4,  pp.  251-­‐259,  03  2017.  

[11]   R.  Kayed  et  al.,  "Conformation  dependent  monoclonal  antibodies  distinguish  different  replicating  strains  or  conformers  of  prefibrillar  Aβ  oligomers,"  (in  eng),  Mol  Neurodegener,  vol.  5,  p.  57,  2010.  

[12]   O.  El-­‐Agnaf  et  al.,  "Differential  effects  of  immunotherapy  with  antibodies  targeting  α-­‐synuclein  oligomers  and  fibrils  in  a  transgenic  model  of  synucleinopathy,"  (in  eng),  Neurobiol  Dis,  vol.  104,  pp.  85-­‐96,  Aug  2017.  

[13]   T.  Tokuda  et  al.,  "Detection  of  elevated  levels  of  α-­‐synuclein  oligomers  in  CSF  from  patients  with  Parkinson  disease,"  (in  eng),  Neurology,  vol.  75,  no.  20,  pp.  1766-­‐72,  Nov  2010.  

[14]   "DATATOP:  a  multicenter  controlled  clinical  trial  in  early  Parkinson's  disease.  Parkinson  Study  Group,"  (in  eng),  Arch  Neurol,  vol.  46,  no.  10,  pp.  1052-­‐60,  Oct  1989.  

[15]   S.  Moussaud,  D.  R.  Jones,  E.  L.  Moussaud-­‐Lamodière,  M.  Delenclos,  O.  A.  Ross,  and  P.  J.  McLean,  "Alpha-­‐synuclein  and  tau:  teammates  in  neurodegeneration?,"  (in  eng),  Mol  Neurodegener,  vol.  9,  p.  43,  2014.  

  159  

[16]   B.  Mollenhauer,  J.  J.  Locascio,  W.  Schulz-­‐Schaeffer,  F.  Sixel-­‐Döring,  C.  Trenkwalder,  and  M.  G.  Schlossmacher,  "α-­‐Synuclein  and  tau  concentrations  in  cerebrospinal  fluid  of  patients  presenting  with  parkinsonism:  a  cohort  study,"  (in  eng),  Lancet  Neurol,  vol.  10,  no.  3,  pp.  230-­‐40,  Mar  2011.  

[17]   L.  Parnetti  et  al.,  "Cerebrospinal  fluid  biomarkers  in  Parkinson's  disease  with  dementia  and  dementia  with  Lewy  bodies,"  (in  eng),  Biol  Psychiatry,  vol.  64,  no.  10,  pp.  850-­‐5,  Nov  2008.  

[18]   I.  van  Steenoven  et  al.,  "α-­‐Synuclein  species  as  potential  cerebrospinal  fluid  biomarkers  for  dementia  with  lewy  bodies,"  (in  eng),  Mov  Disord,  Nov  2018.  

[19]   A.  Korff,  C.  Liu,  C.  Ginghina,  M.  Shi,  J.  Zhang,  and  A.  s.  D.  N.  Initiative,  "α-­‐Synuclein  in  cerebrospinal  fluid  of  Alzheimer's  disease  and  mild  cognitive  impairment,"  (in  eng),  J  Alzheimers  Dis,  vol.  36,  no.  4,  pp.  679-­‐88,  2013.  

[20]   S.  Hall  et  al.,  "Longitudinal  Measurements  of  Cerebrospinal  Fluid  Biomarkers  in  Parkinson's  Disease,"  (in  ENG),  Mov  Disord,  Feb  2016.  

[21]   A.  W.  Schmid,  B.  Fauvet,  M.  Moniatte,  and  H.  A.  Lashuel,  "Alpha-­‐synuclein  post-­‐translational  modifications  as  potential  biomarkers  for  Parkinson's  disease  and  other  synucleinopathies,"  (in  ENG),  Mol  Cell  Proteomics,  Aug  2013.  

[22]   S.  Tenreiro,  K.  Eckermann,  and  T.  F.  Outeiro,  "Protein  phosphorylation  in  neurodegeneration:  friend  or  foe?,"  (in  eng),  Front  Mol  Neurosci,  vol.  7,  p.  42,  2014.  

[23]   A.  Oueslati,  "Implication  of  Alpha-­‐Synuclein  Phosphorylation  at  S129  in  Synucleinopathies:  What  Have  We  Learned  in  the  Last  Decade?,"  (in  eng),  J  Parkinsons  Dis,  vol.  6,  no.  1,  pp.  39-­‐51,  2016.  

[24]   H.  Fujiwara  et  al.,  "alpha-­‐Synuclein  is  phosphorylated  in  synucleinopathy  lesions,"  (in  eng),  Nat  Cell  Biol,  vol.  4,  no.  2,  pp.  160-­‐4,  Feb  2002.  

[25]   N.  K.  Majbour  et  al.,  "Longitudinal  changes  in  CSF  alpha-­‐synuclein  species  reflect  Parkinson's  disease  progression,"  (in  ENG),  Mov  Disord,  vol.  31,  no.  10,  pp.  1535-­‐1542,  Oct  2016.  

[26]   T.  Stewart  et  al.,  "Phosphorylated  α-­‐synuclein  in  Parkinson's  disease:  correlation  depends  on  disease  severity,"  (in  eng),  Acta  Neuropathol  Commun,  vol.  3,  p.  7,  2015.  

[27]   F.  Tateno,  R.  Sakakibara,  T.  Kawai,  M.  Kishi,  and  T.  Murano,  "Alpha-­‐synuclein  in  the  cerebrospinal  fluid  differentiates  synucleinopathies  (Parkinson  Disease,  dementia  with  Lewy  bodies,  multiple  system  atrophy)  from  Alzheimer  disease,"  (in  eng),  Alzheimer  Dis  Assoc  Disord,  vol.  26,  no.  3,  pp.  213-­‐6,  2012  Jul-­‐Sep  2012.  

[28]   J.  B.  Toledo,  A.  Korff,  L.  M.  Shaw,  J.  Q.  Trojanowski,  and  J.  Zhang,  "CSF  α-­‐synuclein  improves  diagnostic  and  prognostic  performance  of  CSF  tau  and  Aβ  in  Alzheimer's  disease,"  (in  eng),  Acta  Neuropathol,  vol.  126,  no.  5,  pp.  683-­‐97,  Nov  2013.  

[29]   A.  Schrag,  U.  F.  Siddiqui,  Z.  Anastasiou,  D.  Weintraub,  and  J.  M.  Schott,  "Clinical  variables  and  biomarkers  in  prediction  of  cognitive  impairment  in  patients  with  newly  diagnosed  Parkinson's  disease:  a  cohort  study,"  (in  eng),  Lancet  Neurol,  vol.  16,  no.  1,  pp.  66-­‐75,  Jan  2017.  

[30]   B.  Mollenhauer  et  al.,  "A  user's  guide  for  α-­‐synuclein  biomarker  studies  in  biological  fluids:  Perianalytical  considerations,"  (in  eng),  Mov  Disord,  vol.  32,  no.  8,  pp.  1117-­‐1130,  Aug  2017.  

  160  

[31]   K.  Del  Tredici,  C.  H.  Hawkes,  E.  Ghebremedhin,  and  H.  Braak,  "Lewy  pathology  in  the  submandibular  gland  of  individuals  with  incidental  Lewy  body  disease  and  sporadic  Parkinson's  disease,"  (in  eng),  Acta  Neuropathol,  vol.  119,  no.  6,  pp.  703-­‐13,  Jun  2010.  

[32]   V.  Donadio  et  al.,  "A  new  potential  biomarker  for  dementia  with  Lewy  bodies:  Skin  nerve  α-­‐synuclein  deposits,"  (in  eng),  Neurology,  vol.  89,  no.  4,  pp.  318-­‐326,  Jul  2017.  

   

                                                     

  161  

List  of  Publications  (h  index  =  10)    

1. Alpha-­‐synuclein  species  as  potential  CSF  biomarkers  for  Dementia  with  Lewy  bodies  Inger  van  Steenoven,  Nour  K.  Majbour,  Nishant  Vaikath,  Henk  W.  Berendse,  MD,  Wiesje  M.  van  der  Flier,  Wilma  D.J.  van  de  Berg,  Charlotte  E.  Teunissen,  Afina  W.  Lemstra,  and  Omar  M.A.  El-­‐Agnaf.    Movement  Disorders.  

2. Duffy  MF,  Collier  TJ,  Patterson  JR,  Kemp  CJ,  Luk  KC,  Tansey  MG,  Paumier  KL,  Kanaan  NM,  Fischer  DL,  Polinski  NK,  Barth  OL,  Howe  JW,  Vaikath  NN,  Majbour  NK,  El-­‐Agnaf  OMA,  Sortwell  CE.  Lewy  body-­‐like  alpha-­‐synuclein  inclusions  trigger  reactive  microgliosis  prior  to  nigral  degeneration.  J  Neuroinflammation.  2018  May  1;15(1):129.  doi:  10.1186/s12974-­‐018-­‐1171-­‐z.  Erratum  in:  J  Neuroinflammation.  2018  May  29;15(1):169.  

3. El-­‐Agnaf  O,  Overk  C,  Rockenstein  E,  Mante  M,  Florio  J,  Adame  A,  Vaikath  N,  Majbour  N,  Lee  SJ,  Kim  C,  Masliah  E,  Rissman  RA.  Differential  effects  of  immunotherapy  with  antibodies  targeting  α-­‐synuclein  oligomers  and  fibrils  in  a  transgenic  model  of  synucleinopathy.  Neurobiol  Dis.  2017  Aug;104:85-­‐96.  doi:  10.1016/j.nbd.2017.05.002.  

 4. Tomlinson  JJ,  Shutinoski  B,  Dong  L,  Meng  F,  Elleithy  D,  Lengacher  NA,  Nguyen  AP,  Cron  GO,  

Jiang  Q,  Roberson  ED,  Nussbaum  RL,  Majbour  NK,  El-­‐Agnaf  OM,  Bennett  SA,  Lagace  DC,  Woulfe  JM,  Sad  S,  Brown  EG,  Schlossmacher  MG.  Holocranohistochemistry  enables  the  visualization  of  α-­‐synuclein  expression  in  the  murine  olfactory  system  and  discovery  of  its  systemic  anti-­‐microbial  effects.  J  Neural  Transm  (Vienna).  2017  Jun;124(6):721-­‐738.  doi:  10.1007/s00702-­‐017-­‐1726-­‐7.    

 5. Majbour  NK,  Chiasserini  D,  Vaikath  NN,  Eusebi  P,  Tokuda  T,  van  de  Berg  W,  Parnetti  L,  

Calabresi  P,  El-­‐Agnaf  OM.  Increased  levels  of  CSF  total  but  not  oligomeric  or  phosphorylated  forms  of  alpha-­‐synuclein  in  patients  diagnosed  with  probable  Alzheimer's  disease.  Sci  Rep.  2017  Jan  10;7:40263.  doi:  10.1038/srep40263.  

 6. Majbour  NK,  El-­‐Agnaf  O.  Cognitive  impairment  in  Parkinson's  disease.  

Lancet  Neurol.  2017  Jan;16(1):23-­‐24.  doi:  10.1016/S1474-­‐4422(16)30329-­‐5.  No  abstract  available.    

7. Landeck  N,  Hall  H,  Ardah  MT,  Majbour  NK,  El-­‐Agnaf  OM,  Halliday  G,  Kirik  D.  A  novel  multiplex  assay  for  simultaneous  quantification  of  total  and  S129  phosphorylated  human  alpha-­‐synuclein.  Mol  Neurodegener.  2016  Aug  22;11(1):61.  doi:  10.1186/s13024-­‐016-­‐0125-­‐0.    

8. Majbour  NK,  Vaikath  NN,  Eusebi  P,  Chiasserini  D,  Ardah  M,  Varghese  S,  Haque  ME,  Tokuda  T,  Auinger  P,  Calabresi  P,  Parnetti  L,  El-­‐Agnaf  OM.  Longitudinal  changes  in  CSF  alpha-­‐synuclein  species  reflect  Parkinson's  disease  progression.  Mov  Disord.  2016  Oct;31(10):1535-­‐1542.  doi:  10.1002/mds.26754.  

 9. Majbour  NK,  Vaikath  NN,  van  Dijk  KD,  Ardah  MT,  Varghese  S,  Vesterager  LB,  Montezinho  LP,  

Poole  S,  Safieh-­‐Garabedian  B,  Tokuda  T,  Teunissen  CE,  Berendse  HW,  van  de  Berg  WD,  El-­‐Agnaf  OM.  Oligomeric  and  phosphorylated  alpha-­‐synuclein  as  potential  CSF  biomarkers  for  Parkinson's  disease.  Mol  Neurodegener.  2016  Jan  19;11(1):7.  doi:  10.1186/s13024-­‐016-­‐0072-­‐9.  

 

  162  

   

   

     

10. Helwig  M,  Klinkenberg  M,  Rusconi  R,  Musgrove  RE,  Majbour  NK,  El-­‐Agnaf  OM,  Ulusoy  A,  Di  Monte  DA.  Brain  propagation  of  transduced  α-­‐synuclein  involves  non-­‐fibrillar  protein  species  and  is  enhanced  in  α-­‐synuclein  null  mice.  Brain.  2015  Dec  30.  pii:  awv376.    

 11. Javed  H,  Menon  SA,  Al-­‐Mansoori  KM,  Al-­‐Wandi  A,  Majbour  NK,  Ardah  MT,  Varghese  S,  

Vaikath  NN,  Haque  ME,  Azzouz  M,  El-­‐Agnaf  OM.  Development  of  Nonviral  Vectors  Targeting  the  Brain  as  a  Therapeutic  Approach  For  Parkinson's  Disease  and  Other  Brain  Disorders.  Mol  Ther.  2015  Dec  24.  doi:  10.1038/mt.2015.232.    

 12. Vaikath  NN,  Majbour  NK,  Paleologou  KE,  Ardah  MT,  van  Dam  E,  van  de  Berg  WD,  Forrest  SL,  

Parkkinen  L,  Gai  WP,  Hattori  N,  Takanashi  M,  Lee  SJ,  Mann  DM,  Imai  Y,  Halliday  GM,  Li  JY,  El-­‐Agnaf  OM.  Generation  and  characterization  of  novel  conformation-­‐specific  monoclonal  antibodies  for  α-­‐synuclein  pathology.  Neurobiol  Dis.  2015  Jul;79:81-­‐99.  doi:  10.1016/j.nbd.2015.04.009.  Epub  2015  Apr  30.  

 13. Buck  K,  Landeck  N,  Ulusoy  A,  Majbour  NK,  El-­‐Agnaf  OM,  Kirik  D.  Ser129  phosphorylation  of  

endogenous  α-­‐synuclein  induced  by  overexpression  of  polo-­‐like  kinases  2  and  3  in  nigral  dopamine  neurons  is  not  detrimental  to  their  survival  and  function.  Neurobiol  Dis.  2015  Jun;78:100-­‐14.  doi:  10.1016/j.nbd.2015.03.008.  Epub  2015  Mar  25.    

14. Jan  O.  Aasly,  Krisztina  K.  Johansen,  Gunnar  Brønstad,  Bjørg  J.  Warø,  Shiji  Varghese,  Nour  K.  Majbour,  Fatimah  Alzahmi,  Katerina  E.  Paleologou,  Dena  A.  M.  Amer,  Abdulmonem  Al-­‐Hayani  and  Omar  M.  A.  El-­‐Agnaf.  Elevated  Levels  of  CSF  α‑Synuclein  Oligomers  in  Healthy  Asymptomatic  LRRK2  Mutation  Carriers.  Front  Aging  Neurosci.  2014  Sep  25;6:248.  doi:  10.3389/fnagi.2014.00248.    

 15. Oskar  Hansson,  Sara  Hall,  Annika  Öhrfelt,  Henrik  Zetterberg,  Kaj  Blennow,  Lennart  Minthon,  

Katarina  Nägga,  Elisabet  Londos,  Shiji  Varghese,  Nour  K  Majbour,  Abdulmonem  Al-­‐Hayani  and  Omar  MA  El-­‐Agnaf.  Levels  of  cerebrospinal  fluid  alpha-­‐synuclein  oligomers  are  increased  in  Parkinson's  disease  with  dementia  and  dementia  with  Lewy  bodies  compared  to  Alzheimer's  disease.    Alzheimer's  Research  &  Therapy  2014,  May  20;  6:25.    

 16. Lucilla  Parnetti,  Lucia  Farotti,  Paolo  Eusebi1,  Davide  Chiasserini,  Claudia  DeCarlo,  David  

Giannandrea,  Nicola  Salvadori,  Viviana  Lisetti,  NicolaTambasco,  Aroldo  Rossi,  Nour  K.  Majbour,  Omar  El-­‐Agnaf  and  Paolo  Calabresi.  Differential  role  of  CSF  alpha-­‐synuclein  species,  tau,  and  Aβ42  in  Parkinson’s  Diseas.  Front  Aging  Neurosci.  2014  Mar  31;  6:53.  doi:  10.3389.  

 

  163  

WORD OF THANKS  

My deepest gratitude goes to my family for their unconditional love and support.

It’s been a privilege to have worked under the direction of Prof. Omar El-Agnaf, Dr.

Wilma van de Berg and Prof. dr. Henk Berendse. Their sincere mentorship and unwavering

support have immensely leveraged the success of this research.

Omar El-Agnaf, I learnt from you the value of knowledge, courage and integrity. And through

the example you set, I came to appreciate the power of dedication, diligence and resilience. It’s

been eight years now since I joined your team. A journey that has transformed my life and

molded the person I’m today. Thank you very much for crafting an inspiring example as a

researcher, mentor, visionary leader and a role model.

Henk Berendse, I have always enjoyed and appreciated your sincere and wise advice. Your

enlightening guidance and constructive feedback have positively influenced my research career.

Wilma van de Berg, my sincere gratitude to you for creating and facilitating this wonderful

opportunity for me. Thank you very much for your kind acts of care, genuine encouragement and

the excellent research experience.

To all my academic educators, thank you very much for the untiring support and

guidance throughout my journey.

To the reading and examination committee, thank you very much for sharing your

time, expert opinion and extensive knowledge.

  164  

Special thanks go to all my colleagues, namely Nishant Vaikath, Mercy Thomas,

Muneera Fayyad, Houari Abdesselem, Ilham Abdi, Yasmine Fathy, Hanneke Guet, Angela

Ingrassia and Tim Moors for their companionship, kind support and the friendly working

atmosphere.

Nishant Vaikath, you have been a wonderful colleague, true mentor and a very

supportive friend.

Warm thanks to my dear friends for helping me unlock the strength I didn’t know I have.

My deepest appreciation for the patients and the healthy volunteers whom research is

not possible without their contribution.

I would also like to extend my gratitude to the VU University Medical Center,

Amsterdam Neuroscience, Amsterdam, the Netherlands for this life-changing experience, the

access to world-class research facilities and the expert mentorship.

                               

  165  

ABOUT THE AUTHOR  Nour Majbour was born 12 March 1987 in Abu Dhabi, United Arab

Emirates for Syrian Parents. After completing her high school

education, she pursued a bachelor in Pharmacy from Tishreen

University at her hometown, Lattakia, Syria. In 2014 she obtained her

Master degree in Biomedical Sciences, from the College of Medicine

and health Sciences, at UAE University. In 2015, Majbour commenced

her PhD project, supervised by Prof. Dr. Henk Berendse, Prof. Omar

El-Agnaf and Dr. Wilma van de Berg at the department of Anatomy and Neurosciences of the VU

University Medical Center, which resulted in the publication of this thesis.

In 2017, Majbour joined Hamad Bin Khalifa University’s (HBKU) Qatar Biomedical Research

Institute (QBRI) as a research associate. One year later and through Stars of Science TV show,

the Arab World’s leading science-based innovation show, Majbour took her research from QBRI’s

laboratories to a global television audience and skillfully fought her way through prototype

performance, industrial design and customer validation with her proposed innovation for early

detection of Parkinson’s disease earning the 2nd place. Majbour hopes her innovation, Parkinson’s

Early Detection Kit (QABY) would surpass the current limitations in disease diagnostics through

the use of novel antibodies that may be leveraged to identify biomarkers for Parkinson’s disease.

Looking towards the marketability of her innovation, Majbour plans to use her wining share of

seed money to develop her kit for an up-and-coming Qatari biotech company that aims to develop

new scientific tools for early diagnosis, treatment, and management of devastating diseases, thus

supporting Qatar’s position as a global player in the healthcare industry. Her entrepreneurial spirit

and unwavering dedication, which stem from personal experiences with family members afflicted

by Parkinson’s disease, positioned her as a strong contestant and effective role model for women

in Science, Technology, Engineering and Mathematics (STEM) professions.

 

 

Department of Anatomy and Neurosciences