Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121,...

19
FORUM REVIEW ARTICLE Causes and Consequences of Degeneration of the Dorsal Motor Nucleus of the Vagus Nerve in Parkinson’s Disease James G. Greene Abstract Significance: Parkinson’s disease (PD) is no longer considered merely a movement disorder caused by de- generation of dopamine neurons in the midbrain. It is now recognized as a widespread neuropathological syndrome accompanied by a variety of motor and nonmotor clinical symptoms. As such, any hypothesis concerning PD pathogenesis and pathophysiology must account for the entire spectrum of disease and not solely focus on the dopamine system. Recent Advances: Based on its anatomy and the intrinsic properties of its neurons, the dorsal motor nucleus of the vagus nerve (DMV) is uniquely vulnerable to damage from PD. Fibers in the vagus nerve course throughout the gastrointestinal (GI) tract to and from the brainstem forming a close link between the peripheral and central nervous systems and a point of proximal contact between the environment and areas where PD pathology is believed to start. In addition, DMV neurons are under high levels of oxidative stress due to their high level of a-synuclein expression, fragile axons, and specific neuronal physiology. Moreover, several consequences of DMV damage, namely, GI dysfunction and unrestrained inflammation, may propagate a vicious cycle of injury affecting vulnerable brain regions. Critical Issues: Current evidence to suggest the vagal system plays a pivotal role in PD pathogenesis is circumstantial, but given the current state of the field, the time is ripe to obtain direct experimental evidence to better delineate it. Future Directions: Better understanding of the DMV and vagus nerve may provide insight into PD pathogenesis and a neural highway with direct brain access that could be harnessed for novel therapeutic interventions. Antioxid. Redox Signal. 00, 000–000. Introduction P arkinson’s disease (PD) is a common neurological degenerative disorder affecting 1% of individuals over 55 years of age, the cardinal features of which are related to motor function: resting tremor, rigidity or stiffness, brady- kinesia or slowness of movement, and postural instability (71). Additionally, PD is associated with a multitude of nonmotor symptoms, including disturbances of sleep, cog- nition, mood, autonomic regulation, and gastrointestinal (GI) function (230). As therapy for the motor features of PD has advanced dramatically, treatments of the nonmotor symp- toms of the disorder have lagged behind to the point that they have become some of the most common and frustrating problems faced by PD patients (230). For example, more than 8/10 hospital admissions for PD are directly due to nonmotor symptoms, and the majority of disability in later stage PD is related to symptoms that do not respond to levodopa, the most effective motor therapy for PD (128, 266, 287). Much as the recent understanding of PD as a clinical syndrome broadened from one focused narrowly on motor symptoms, so has the understanding of PD as a neuropatho- logical syndrome broadened from one focused on dopamine neurons in the midbrain substantia nigra to one encompassing widespread regions of the central nervous systems (CNS) and peripheral nervous systems (PNS) (16, 17, 35, 159). In ad- dition to the substantia nigra, neuronal loss has been con- vincingly described for noradrenergic neurons in the pontine locus ceruleus, cholinergic and noradrenergic neurons in sympathetic centers in the spinal cord and periphery, and cholinergic neurons in the dorsal motor nucleus of the vagus nerve (DMV) in the medulla, among other areas (19, 86, 95, 96, 103, 104, 119, 131, 209). Furthermore, the pathological hallmark of PD, Lewy bodies as detected by abnormal a- synuclein (AS) immunoreactivity, is spread through an even wider variety of neurochemical subtypes, including cells in the olfactory bulb, cerebral cortex, pontine raphe, spinal cord, and enteric nervous system (ENS) (16, 35, 77, 139, 165). Department of Neurology, Emory University, Atlanta, Georgia. ANTIOXIDANTS & REDOX SIGNALING Volume 00, Number 00, 2014 ª Mary Ann Liebert, Inc. DOI: 10.1089/ars.2014.5859 1

Transcript of Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121,...

Page 1: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

FORUM REVIEW ARTICLE

Causes and Consequences of Degeneration of the DorsalMotor Nucleus of the Vagus Nerve in Parkinson’s Disease

James G. Greene

Abstract

Significance: Parkinson’s disease (PD) is no longer considered merely a movement disorder caused by de-generation of dopamine neurons in the midbrain. It is now recognized as a widespread neuropathologicalsyndrome accompanied by a variety of motor and nonmotor clinical symptoms. As such, any hypothesisconcerning PD pathogenesis and pathophysiology must account for the entire spectrum of disease and not solelyfocus on the dopamine system. Recent Advances: Based on its anatomy and the intrinsic properties of itsneurons, the dorsal motor nucleus of the vagus nerve (DMV) is uniquely vulnerable to damage from PD. Fibersin the vagus nerve course throughout the gastrointestinal (GI) tract to and from the brainstem forming a close linkbetween the peripheral and central nervous systems and a point of proximal contact between the environment andareas where PD pathology is believed to start. In addition, DMV neurons are under high levels of oxidative stressdue to their high level of a-synuclein expression, fragile axons, and specific neuronal physiology. Moreover,several consequences of DMV damage, namely, GI dysfunction and unrestrained inflammation, may propagate avicious cycle of injury affecting vulnerable brain regions. Critical Issues: Current evidence to suggest the vagalsystem plays a pivotal role in PD pathogenesis is circumstantial, but given the current state of the field, the timeis ripe to obtain direct experimental evidence to better delineate it. Future Directions: Better understanding ofthe DMV and vagus nerve may provide insight into PD pathogenesis and a neural highway with direct brainaccess that could be harnessed for novel therapeutic interventions. Antioxid. Redox Signal. 00, 000–000.

Introduction

Parkinson’s disease (PD) is a common neurologicaldegenerative disorder affecting 1% of individuals over 55

years of age, the cardinal features of which are related tomotor function: resting tremor, rigidity or stiffness, brady-kinesia or slowness of movement, and postural instability(71). Additionally, PD is associated with a multitude ofnonmotor symptoms, including disturbances of sleep, cog-nition, mood, autonomic regulation, and gastrointestinal (GI)function (230). As therapy for the motor features of PD hasadvanced dramatically, treatments of the nonmotor symp-toms of the disorder have lagged behind to the point that theyhave become some of the most common and frustratingproblems faced by PD patients (230). For example, more than8/10 hospital admissions for PD are directly due to nonmotorsymptoms, and the majority of disability in later stage PD isrelated to symptoms that do not respond to levodopa, the mosteffective motor therapy for PD (128, 266, 287).

Much as the recent understanding of PD as a clinicalsyndrome broadened from one focused narrowly on motorsymptoms, so has the understanding of PD as a neuropatho-logical syndrome broadened from one focused on dopamineneurons in the midbrain substantia nigra to one encompassingwidespread regions of the central nervous systems (CNS) andperipheral nervous systems (PNS) (16, 17, 35, 159). In ad-dition to the substantia nigra, neuronal loss has been con-vincingly described for noradrenergic neurons in the pontinelocus ceruleus, cholinergic and noradrenergic neurons insympathetic centers in the spinal cord and periphery, andcholinergic neurons in the dorsal motor nucleus of the vagusnerve (DMV) in the medulla, among other areas (19, 86, 95,96, 103, 104, 119, 131, 209). Furthermore, the pathologicalhallmark of PD, Lewy bodies as detected by abnormal a-synuclein (AS) immunoreactivity, is spread through an evenwider variety of neurochemical subtypes, including cells inthe olfactory bulb, cerebral cortex, pontine raphe, spinal cord,and enteric nervous system (ENS) (16, 35, 77, 139, 165).

Department of Neurology, Emory University, Atlanta, Georgia.

ANTIOXIDANTS & REDOX SIGNALINGVolume 00, Number 00, 2014ª Mary Ann Liebert, Inc.DOI: 10.1089/ars.2014.5859

1

Page 2: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

Thus far, data indicate that many of these areas are morevulnerable than the midbrain dopamine neurons to Parkin-sonian pathology and death. In fact, the extreme focus onpathology in dopamine neurons over the past century mayhave limited our understanding of PD as a more systemicneurological disorder (159, 169, 261).

This view of PD is more a rediscovery than a new one.When James Parkinson first described the disorder in 1817,he described a multitude of nonmotor symptoms and hy-pothesized that the ‘‘proximate cause’’ was a ‘‘diseased stateof the medulla spinalis.extending, as the disease proceeds,to the medulla oblongata (216).’’ Frederic Lewy describedthe pathological hallmark of PD, the Lewy body, in the early1900s in the basal forebrain and medulla before noting themin the midbrain (172, 173). As the field has returned to con-sidering PD as a systemic disease, many theories concerningits pathophysiology focused solely on dopamine neuronshave required reevaluation. Although the dopaminergicphenotype is still an important component related to selectiveneuronal vulnerability in PD, it is not the only one. In par-ticular, anatomical and pathophysiological links between thePNS and CNS and the specific subtypes of affected neuronswithin each are prerequisites to any comprehensive theory ofPD pathogenesis.

The DMV and the vagus nerve provide such a link. Lo-cated in the lower brainstem, the DMV contains widelyramified neurons projecting to nearly every part of the pe-riphery. In addition, the vagal complex has extensive inter-connections to the rest of the brain. Stimulation of the nervehas profound effects in both central and peripheral directionswith the vagal nerve stimulation widely used as a treatmentfor epilepsy and depression refractory to medications andexperimentally used for its cardioprotective, antiasthmatic,anti-inflammatory, and GI promotility effects (32, 59, 116,154, 181, 245, 271).

In this review, I will review the basic anatomy of the DMV,discuss the impact of PD on the nucleus, and review potentialconsequences of and causes for DMV degeneration in PD.

Functional Anatomy of the Vagus Nerve and DMV

The vagus nerve is a mixed cranial nerve (the 10th) thatcontains (in descending order of abundance) (i) afferentvisceral sensory (predominantly autonomic) fibers from theheart, lungs, GI tract and accessory organs (spleen, liver,gallbladder), sweat glands, and other areas; (ii) efferent vis-ceral motor (autonomic) fibers to a roughly similar distribu-tion of tissues; (iii) efferent somatic motor fibers to severalskeletal muscles of the pharynx and larynx; (iv) afferentspecial sensory fibers mediating taste in portions of thepharynx; and (v) afferent general sensory fibers from parts ofthe ear and surrounding skin (Fig. 1) (234).

Vagus nerve functions are regulated by a central auto-nomic network that integrates visceroceptive, humoral, andenvironmental information and modulates autonomic, endo-crine, behavioral motor, emotional, attentional, and anti-nociceptive output (18, 45, 182). The structures forming thecentral autonomic network are widely distributed at all levelsof the nervous system, including the cerebral cortex, basalforebrain, basal ganglia, midbrain, thalamus, hypothalamus,pons, and medulla (45, 46, 180, 202). Although less well-studied than for motor function, the basal ganglia appear toplay a similar comparator role in autonomic function mod-ulating the iterative processing of the central network; bothanatomical and neurophysiological connections have beendescribed (5, 54, 67, 90, 92, 105, 112, 118, 138, 158, 176,178, 183, 223–225, 259, 264).

The DMV is a paired nucleus on each side of the caudalmedulla that runs longitudinally along the dorsomedial as-pect of the brainstem (Fig. 1). It gives rise to preganglionic

FIG. 1. Anatomy of the DMV and vagus nerve. (A) Schematic of a ventral view of the human brainstem noting theapproximate location of the DMV. (B) Schematic of a cross section of the human brainstem at the level of the medullaindicating different components of the vagus nerve. DMV, dorsal motor nucleus of the vagus nerve; GI, gastrointestinal;NTS, nucleus of the tractus solitarius; Nuc., nucleus; n., nerve.

2 GREENE

Page 3: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

parasympathetic autonomic fibers that innervate the GI tractand accessory organs from the distal third of the esophagus toapproximately the splenic flexure of the colon, although thereis some evidence for more distal projections (22). Cardiovas-cular preganglionic parasympathetics arise predominantlyfrom the nucleus ambiguus rather than the DMV (206).

DMV neurons have thin, widely ramified axons with min-imal myelination that are among the longest in the body (33,169). This is in contradistinction to other fibers in the vagusnerve (afferent and somatic motor efferent) with adjacent lo-cations in the medulla that have more robustly myelinatedneurites with simpler branching structures. Acetylcholine isthe major neurotransmitter for DMV neurons, although othersare thought to function as cotransmitters, including substanceP and catecholamines.

Parasympathetic innervation from the DMV is importantfor control of GI motility and reflexes, especially in thestomach (219, 237, 267, 268). About 25%–30% of fibers inthe abdominal vagus nerve are efferent fibers that modulatefunction of the ENS, a semi-autonomous network of neuronslining the entire alimentary canal to control GI function(142). Individual axons from DMV neurons ramify widelyinto an extensive network contacting nearly every entericneuron, especially in the proximal GI tract (Fig. 2) (22, 205).Vagotomy has long been known to result in GI dysfunction;less well known are GI complications due to pathologicallesions of the DMV itself (240, 241, 254).

In addition to its classical function as a modulator of GIfunction, the DMV has recently been described as a potent in-hibitor of inflammation via the cholinergic anti-inflammatorypathway (32, 248, 279). Studies have shown that activation ofvagal parasympathetics attenuates the systemic inflammatoryresponse to a variety of insults, including endotoxin admin-istration (32). This effect is mediated via the a7 subunit ofthe nicotinic acetylcholine receptor, which is expressed onmacrophages (279). In addition to the anti-inflammatory ef-fect of vagus nerve stimulation, physical or pharmacologicalvagotomy has a proinflammatory effect, in which the branchof the vagus nerve that innervates the spleen has been shownto be particularly important (137, 222, 246). The afferent

signaling for this iterative inflammatory/anti-inflammatoryloop has both neural and humoral components and is mod-ulated by multiple central and systemic signaling mecha-nisms (76, 208, 220, 221, 247, 248, 288, 289).

Pathophysiology of the DMV in PD

There are two main ways that PD may affect the vagalsystem. One is the direct impact of neuronal damage to vagusneurons. For example, loss of DMV innervation to the gutcauses GI problems by cutting a hard-wired pathway. Theother is the downstream deleterious effect of central neuro-degeneration on iterative processing of autonomic regulation.For example, from a motor standpoint, it is now accepted thatmotor parkinsonism is a circuit disorder, rather than a straightline from dopamine depletion to motor dysfunction. Given theevidence accumulated thus far, autonomic symptoms in PDmay be caused by both mechanisms. Whereas this review willprimarily focus on the direct damage accumulated by DMVneurons in PD, considering the DMV as one node in a dis-tributed network is helpful to place these findings in context.

Disrupted central regulation of DMV output

Loss of substantia nigra neurons and subsequent striataldopamine depletion is a primary driver of PD motor symp-toms by disrupting basal ganglia circuit function (39, 49, 61,101, 121, 171, 188, 189, 207, 249, 255). Loss of integrity inthe circuitry of the striatum increases the oscillatory activityin corticocortical loops by propagating to basal ganglia out-put nuclei (63, 69, 102, 187, 239). Experimental studies haveshown that pharmacological blockade of dopamine receptors(61, 87) or lesion of the nigrostriatal pathway (284, 285)increases the neuronal synchronization and oscillatory activityin the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brainstimulation have been found to reduce synchronization inconjunction with their therapeutic effect (70, 274). While thisabnormal circuit activity has been intensively investigated inregard to motor function, its contribution to other symptoms isless clear.

FIG. 2. Anatomy of inter-actions between the vagusnerve and enteric nervoussystem (ENS) in the GI tract.(A) Schematic of intercon-nections between the vagusand ENS. (B) Efferent nerveterminals from the DMV sur-rounding a neuronal ganglionin the myenteric plexus high-lighted by a-synuclein im-munostaining in a transgenicmouse. (C) Neurites in prox-imity to the intestinal lumenhighlighted by peripherin im-munostaining in a mouse(205). Arrows mark a neuritein an intestinal villus and ar-rowheads mark enteric neu-rons in ganglia.

DMV IN PD 3

Page 4: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

The relationship between dopamine depletion and auto-nomic dysfunction has been examined in a few previousstudies. In rats, the salivary response is dependent upon theactivity of dopamine receptors in the striatum and decreasedby striatal lesions (223–225). Baroreflex regulation of bloodpressure has been reported to be dependent on the nigros-triatal pathway in rodents (5, 67, 176, 178, 183). In addition,GI motility can be modulated by basal ganglia activity, es-pecially in the caudate (112, 158, 264). In some PD patients,deep brain stimulation in the subthalamic nucleus has beenreported to improve cardiovascular and GI dysfunction (134,161, 292).

More specific to the vagal system, animals with lesions ofthe nigrostriatal tract show reduced choline acetyltransferaseexpression in the DMV and decreased levels of acetylcholinein the gastric wall (291). Although dopamine receptors areexpressed by both cholinergic and catecholaminergic neu-rons in the DMV (43), a direct anatomical link between thetwo structures has not yet been demonstrated.

It is enticing to hypothesize that there are loop circuitsregulating the autonomic function analogous to those dem-onstrated for somatic motor control and that these loops maynot only link a distributed network of brain nuclei but also theperipheral and ENS. This is likely to be a fruitful area ofresearch in the future; however, at present, the existence anddysfunction of such loops are largely hypothetical, whiledirect pathological damage to the DMV has been clearly andrepeatedly demonstrated in PD patients.

Neuropathology of the DMV in PD

Although PD has predominantly been considered a diseaseof dopaminergic neurons in the substantia nigra, it has longbeen known that there is substantial neuropathology in otherareas. For example, extensive cell loss on the order of > 50%in the DMV has been described consistently in PD since 1925(82, 88, 126). This loss is age dependent in PD in that, olderage correlates with fewer DMV neurons; however, there is noage-dependent loss of DMV neurons in individuals withoutPD (95). Preganglionic parasympathetic neurons in the DMVare preferentially vulnerable to degeneration (82, 119, 120).

Lewy bodies, eosinophilic proteinaceous intracellular in-clusions that are a pathological hallmark of PD, were de-scribed in the DMV before their recognition in the substantianigra (172, 173, 226). Additionally, the DMV has been notedto have the highest number and density of ubiquitin-positivedegenerating neurites of any brain nucleus in PD. Intrigu-ingly, the density of degenerating neurites in the DMV cor-relates with the duration of PD, while the density in thesubstantia nigra does not (94).

In 1997, mutations in a-synuclein (AS) were described asan autosomal dominant cause of parkinsonism (231). Soonafter, it was described that AS was a major component ofLewy bodies in all cases of idiopathic PD (260). Thesefindings led to a renewed appreciation for Lewy pathologyand careful examination of its extent and character in PD.Using immunostaining techniques, AS aggregation, particu-larly in neurites, has been shown to be very prominent in theDMV (35, 145, 217).

Lewy neurites have been described in the ENS in nearly100% of PD patients at autopsy (Fig. 3) (6, 155, 236, 275–277). The vast majority ( > 97%) of ENS synuclein pathology

in PD is not cytoplasmic Lewy bodies in enteric neuron cellbodies, but neuritic Lewy pathology around the ENS, afinding that is recapitulated in AS transgenic mice (34, 276,277). The distribution of pathology in both humans andtransgenic mice mirrors that of efferent DMV innervation tothe ENS nearly exactly, where neuritic AS aggregation andLewy pathology are common proximally (stomach) and raredistally (rectum) (6, 17, 205, 276, 277).

It has been suggested that AS neuritic pathology affects theDMV very early in the course of PD, and furthermore, neu-ritic AS pathology in the GI tract occurs at least as early, if notearlier (33–35). In fact, Braak has proposed a pathologicalstaging system of PD based on AS pathology derived fromcharacterization of AS neuritic pathology in brains from PDpatients as well as aged patients where incidental AS neuriticpathology was found on neuropathological examination (33,35). The scheme proposes that PD pathology begins in theDMV, with subsequent spread of AS pathology rostrally fromthe DMV to pontine nuclei (locus ceruleus, raphe), to mid-brain (substantia nigra), and last, to the cortical areas. This

FIG. 3. Examples of Lewy bodies in the GI tract. (A)Lewy body in a myenteric ganglion in the ileum from a PDpatient highlighted by a-synuclein immunostaining. (B)Lewy bodies in the mucosa and submucosa in the stomachfrom a PD patient highlighted by a-synuclein im-munostaining (6). PD, Parkinson’s disease.

4 GREENE

Page 5: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

model is quite controversial, with some authors in agreementand others less so (41, 144, 145, 177, 217). In particular, atheory of progressive pathogenesis based on cross-sectionalpostmortem data and the assumption that patients with inci-dental Lewy pathology would ultimately develop PD isproblematic. However, there is uniform agreement that theDMV is consistently and severely affected by PD pathologyearly in the disease course.

Consequences of DMV Degeneration in PD

Given the functions of the vagus nerve, there are multiplepotential consequences to dysfunction and degeneration ofthe DMV that may impact PD patients, including GI dys-function and unchecked inflammation. In addition to directimpact on patient symptoms and quality of life, damage toDMV neurons could contribute to a feed-forward cycle in thepathophysiology of PD whereby loss of DMV functionworsens neuropathology (Fig. 4).

GI dysfunction

GI dysfunction is a prominent nonmotor symptom in PDthat occurs in nearly every patient at some point in his or herillness (85, 227, 228). Symptoms span the entire alimentarytract and include early satiety and nausea from delayed gas-tric emptying, bloating from poor small bowel coordination,and constipation and defecatory dysfunction from impairedcolonic transit (84, 85, 196, 227).

A comprehensive survey of PD patients in the late 1980’sfound that problems with saliva occurred in 70% of PD pa-tients, dysphagia (trouble swallowing) in 52%, nausea in 24%,and constipation in 29%, whereas the same symptoms werereported by less than 10% of age-matched control individuals.In addition, defecatory dysfunction was reported by two-thirdsof PD patients—twice the control prevalence (84).

Subsequent studies have confirmed the high frequency ofGI abnormalities in PD. Abnormal gastric emptying has beendescribed in 43%–88% of PD patients and can worsen as PDprogresses (79, 106, 107, 125, 153, 201). Interestingly, ob-jective gastric motility abnormalities are frequently detected

even in PD patients without subjective GI complaints (50).The incidence rate of constipation in PD has been estimatedto be anywhere from 29% to 81%, and problems with defe-cation are also much more common in PD (*60%) than inage-matched controls (*25%) (83, 250, 256, 257).

GI dysfunction has many important consequences for PDpatients. First, GI symptoms negatively impact the quality oflife. They have significant psychosocial impact by causingalterations in eating habits, contributing to feelings of em-barrassment, and invoking the need for social and caregiveradjustment (196, 250). Second, GI dysfunction in PD is as-sociated with serious, possibly life-threatening complica-tions, including pulmonary aspiration, weight loss,malnutrition, megacolon, and intestinal pseudo-obstruction(1, 12, 85, 147, 190, 204, 251). Third, GI dysfunction dra-matically impacts motor PD symptoms by causing erraticabsorption of oral medications, motor fluctuation, and med-ication side effects (10, 79, 80, 106, 107, 156, 201). Finally, ithas recently been hypothesized that GI disturbance may be asentinel event in the manifestation of PD. Constipation canappear decades before motor symptoms in PD, and recentdata from a large-scale study suggest that constipation isassociated with an increased risk of future PD (2, 269).

The majority of GI symptoms have objective correlatesrelated to abnormal motility of the GI tract. For example, notonly do patients describe symptoms of early satiety andbloating but also they have evidence that slowed stomachmotility and delayed gastric emptying are responsible forthose symptoms (50, 79, 107). Similarly, patients experi-encing constipation have objective evidence for slowed co-lonic transit, and patients with defecatory dysfunction haveelectrophysiologic sphincter abnormalities (8, 9, 84, 143).The total GI tract transit time is also significantly prolongedin PD (66).

In addition to dysmotility, it has recently been suggestedthat PD patients have increased GI permeability and gutleakiness. Two small studies have indicated using noninva-sive testing that absorption of nonmetabolizable sugars isgreater in PD patients, implying that permeability across theGI mucosa is higher (89, 252). One has shown an increase in

FIG. 4. Damage to the DMV may prop-agate a vicious cycle of neuronal damagein PD. This is a schematic hypothesis of theinvolvement of the DMV in PD pathogenesis.Factors intrinsic and extrinsic to the DMVcontribute to pathology in an iterative cycle.The trigger or starting point for such a cycleis not known.

DMV IN PD 5

Page 6: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

Escherichia coli density in the epithelium and lamina propriaand an indication that PD patients have greater exposure tobacterial endotoxin (89). These findings correlated with ab-normal AS immunoreactivity and markers of neuroin-flammation; however, data are scarce, and the extent andcauses of increased gut permeability are unknown.

Despite the frequency of GI dysfunction and GI neuro-pathology in PD, it is not clear whether or not there is acausal relationship between the two findings, and clinico-pathological correlation studies have only begun to beperformed (6, 72, 89, 164, 165). Recent evidence from co-lon biopsies indicates both synuclein aggregation and neu-ronal loss in the submucosal plexus of the ENS possiblycorrelated with GI symptoms in some way (163, 165, 232).This is in contradistinction to the myenteric plexus of theENS which, although it contains a greater burden of synu-clein pathology, does not exhibit loss of neurons (6). As faras the potential role of vagal pathology, there is a casereport of motor fluctuations first developing in a PD patientimmediately after vagotomy (78). In addition, transgenicanimals expressing abnormal AS in the DMV exhibit age-dependent GI dysfunction similar to that seen in PD in theabsence of synuclein pathology in enteric neurons them-selves (205). Experience from the midbrain suggests thatclinical symptoms in PD are driven primarily by neuronalloss rather than aggregation of a-synuclein, so quantitativeevaluation of neuron populations that control GI motility isan important step toward determining the pathological un-derpinnings of GI symptoms in PD. In this regard, explo-ration of DMV pathology is more advanced than that forother neuronal populations that modulate GI motility sinceneuronal loss has been convincingly described in PD;however, no direct correlations between vagal pathologyand GI symptoms have been completed (82, 88, 126). Ob-viously, these would be difficult studies to perform, sincepathological evaluation of the DMV necessarily occurspostmortem, but organizations such as the Arizona Parkin-son’s Disease Consortium and the Banner Sun Health Re-search Institute Brain and Body Donation Program haverecently begun to address this need (6). Equally intriguing isthe potential to use advanced imaging techniques such asfunctional magnetic resonance imaging to monitor DMVactivity in real time in PD patients or animal models with GIsymptoms (150, 186, 268, 270, 282, 283).

Unchecked inflammatory responses

Activated microglia, accumulation of proinflammatory cy-tokines, nuclear factor kappa B pathway activation, and oxi-dative damage to proteins have all been described in the brainsof individuals with PD (132, 262, 263). Intriguingly, themidbrain from patients or primates exposed to the dopa-mine neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine(MPTP) years earlier (supposedly a one-time event) displayedsimilar microglia activation and signs of active neurodegen-eration at autopsy suggesting that common mechanismsmay underlie degeneration induced by divergent triggers (14,160, 195). In addition, the chronic use of nonsteroidal anti-inflammatory drugs, in particular, ibuprofen, has been asso-ciated with a lower incidence of PD (51, 52, 253).

Experimental evidence from rodent models also supports arole for both innate and adaptive immune system modulation

of dopamine neuron survival. Exposure of dopamine neuronsto the gram-negative bacterial endotoxin lipopolysaccharide,after either direct infusion into the midbrain or intraperitonealinjection, results in delayed loss of dopamine neurons asso-ciated with persistent neuroinflammation in the midbrain (91,98, 99, 235). Even dopaminergic damage caused by nonin-flammatory insults such as MPTP or 6-hydroxydopamine isprevented by blockade of proinflammatory cytokines such astumor necrosis factor a, inhibition of cyclooxygenase, oralteration of adaptive immunity by transfer of T lymphocytes(20, 37, 91, 136, 168, 194, 242, 243, 265). Importantly,transient initiation factors (i.e., toxins, bacterial or viral in-fections, particulate matter, and pesticides) may trigger anactive, self-perpetuating cycle of chronic neuroinflammation(i.e., increased production of chemokines, cytokines, reactiveoxygen species/reactive nitrogen species, and adhesionmolecules by activated microglia), which serves to furtherpromote clustering of activated microglia around dopamineneurons and may contribute to irreversible neuronal dys-function and cell death (13, 38).

Although selective loss of midbrain dopamine neuronsfrom the substantia nigra pars compacta is the typically rec-ognized pathological hallmark of PD, as discussed above,many levels of the nervous system are pathologically affectedby PD, with the DMV, one of the most severely and earliestaffected spots (82, 88, 126). The usual interpretation of thesefindings has been that the same pathological processes thatimpact dopamine neurons affect DMV neurons. That maycertainly be correct; however, the early involvement of theDMV and a broader vagal system may have additional im-plications for PD pathogenesis. In particular, the possibilitythat early loss of DMV neurons and a resultant deficiency ofvagal anti-inflammatory signaling exacerbate damage todopamine neurons is intriguing because if true, it would opena window of opportunity to protect dopamine neurons in PD.Whereas not explored in any neurodegenerative diseases todate, the hypothesis that anti-inflammatory actions mediatedby the vagus nerve could be neuroprotective is gainingtraction in other brain diseases such as stroke and traumaticbrain injury (53, 133, 154). It has been known for years thatpeople who smoke have a lower incidence of PD and thatnicotine is protective in animal model systems (30, 117, 149,233, 238). Given that the anti-inflammatory effects of thevagus are dependent on stimulation of nicotinic acetylcholinereceptors, it is possible that the protective effect of nicotineagainst PD is explained by the anti-inflammatory effects ofnicotinic receptor activation (148, 214).

Causes of DMV Degeneration in PD

There are myriad proposed mechanisms of neurode-generation in PD with the precise one likely a combinationof environmental and host factors, including genetics. Thissection will review proposed causes of neurodegenerationwith a focus on the unique qualities of DMV neurons thatmay make them particularly susceptible to damage in PD. Inaddition, the consequences of damage to DMV neuronsdescribed above are likely contributors to a feed-forwardcycle in the pathophysiology of PD (Fig. 4). A slowerGI transit increases the exposure to potentially toxic en-vironmental agents, increases inflammation by alteringthe luminal microbiotic environment, and disrupts brain-

6 GREENE

Page 7: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

gut feedback loops; increased epithelial permeability hassimilar consequences. For example, although runaway in-flammation may be a consequence of DMV damage, it mayalso be a contributing cause.

Inflammation and neuroinflammation

Although it has not yet been studied, DMV neurons aretheoretically susceptible to many of the same inflammatoryinsults as dopamine neurons. Perhaps, more prescient withregard to the role of inflammation in PD is the unique positionbridging the central and PNS. In fact, DMV neurons arepotentially exposed to a myriad of inflammatory insults viaaxon terminals in proximity to the GI lumen and, as such, theoutside world (Fig. 2). This anatomy has led to the provoc-ative idea that PD may be caused by an unidentified envi-ronmental agent that gains access to the CNS via the GI tractand vagus nerve (36).

The anatomy (or highway) to support such a theory is fairlywell known (193, 198). More intriguingly, mice exposed tothe influenza virus not only show viral infection in serialprogression from the ENS to the DMV to substantia nigradopamine neurons but evidence of AS aggregation, micro-glial activation, and neurodegeneration in the same regions(140). This raises the possibility that a localized, transientevent could propagate in time and space to become a wide-spread self-sustaining neurodegenerative cycle.

A clinical association between the GI tract and inflam-mation in PD is just starting to be investigated, but recentepidemiologic and clinical evidence suggests a link. Pro-inflammatory cytokines and markers of glial activation areupregulated in the colon from PD patients (75). In addition,eradication of Helicobacter pylori (a common cause of gas-tric inflammation and ulcers) modifies PD progression andclinical deterioration (28, 81). Moreover, serum Helicobacterantibody profile may predict the presence, severity, andprogression of PD (281). Recently, a polymorphism in theCARD15 gene previously associated with inflammatory bo-wel disease (IBD) has been associated with PD (26); viceversa, LRRK2, a causative PD mutation, is a susceptibilitylocus for IBD (15). Another putative PD gene, NR4A2, hasalso been linked to GI inflammation (122, 162). In rats, IBDexacerbates in vivo damage to midbrain dopamine neurons byan inflammation-mediated process (273).

As discussed in the next section, recent evidence suggeststhat one potential trigger agent could be aggregated synucleinitself acting via a prion-like mechanism (124). In addition topropagation of pathology by direct transfer, synuclein cantrigger neuroinflammation, causing activation of macrophages,microglia, and astrocytes (3, 167). In fact, neuroinflammationand AS dysfunction can potentiate each other in animal modelsystems and drive chronic neurodegeneration (98, 100).

AS aggregation

Genetic abnormalities in AS cause parkinsonism clinicallyindistinguishable from idiopathic PD; pathological data aremore limited but indicate a similar distribution of Lewy pa-thology and neuronal loss (231, 258). As described above, ASis a major component of Lewy bodies in all cases of PD, andall types of neurons lost in PD display AS-positive Lewypathology (260). The converse is not necessarily true in that itis not clear every neuron containing aggregated AS invari-

ably dies in PD. In addition, the toxic nature of AS is stillbeing debated, and it is possible that aggregation is an attemptat a cytoprotective response rather than a cause of neuronaldeath (7, 60). Regardless of those caveats, hypotheses aboutmechanisms of neurodegeneration in PD must address AS.

The distribution of pathological AS aggregation has beendiscussed above, but AS in its native form is also expressed inDMV neurons. In rodents, AS is expressed at high levels inDMV neurons as compared with other neurons affected in PD(175). In addition, AS is very highly expressed in all efferentaxons from the DMV to the ENS, and there is also expressionof native AS in a proportion of enteric neurons (229). Giventhe widely ramified nature of each DMV axon, the burden ofnative synuclein expression in the GI tract is substantial,particularly in proximal segments like the stomach, whichcorrelates with the distribution of AS neuritic pathologyobserved in PD (6).

The exact pathophysiological mechanisms of neuronaldeath related to AS are unknown, but hypotheses abound.One involves oligomerization, fibrillation, and aggregation,in which native or mutant AS forms toxic multimeric con-structs that disrupt cellular processes (130). Another is thatoverexpression or mutation of AS causes abnormalities inmitochondrial morphology and function (135, 191). Directmitochondrial accumulation of AS with resultant complex Iinhibition has been reported in cellular models, rat brain, andPD brains (74, 179, 212, 213). A third suggests that over-expression of AS disrupts axonal transport and causes mor-phological changes in nerve terminals before cell body loss(56, 166). Another implicates increased generation of toxicoxygen or nitrogen free radical species causing indiscrimi-nate cellular damage or specific activation of downstreamapoptotic pathways (31, 135). Finally, impaired proteindegradation may facilitate all the possible mechanisms listedabove by accelerating accumulation of abnormal, misfolded,or aggregated AS (24). At present, it seems most likely thatall of the above mechanisms interact to form a complicatedinterconnecting pathophysiological network either caused orexacerbated by AS.

Particularly interesting for DMV neurons, whose terminalfields nearly abut the GI lumen, has been the recent findingthat AS and its associated pathology can propagate from cellto cell (124). The question was initially raised after a post-mortem neuropathological evaluation of neuronal transplantsplaced into the striatum of PD patients in the 1980’s revealedAS pathology and neurodegeneration in the transplanted cells(151, 152, 174). Subsequently, it was discovered that AS cantransfer from neuron to neuron and seed pathology in thereceiving neuron by a prion-like mechanism both in vitro andin vivo (73, 123, 192, 199, 200). Pathological forms of AShave been shown to induce Lewy-like pathology in normalmice and even more remarkably, to cause selective degen-eration of dopamine neurons after injection into the striatum(184, 185, 192). This process apparently involves extracel-lular release of AS, perhaps transynaptically, although theprecise mechanisms and location of transfer have not yetbeen conclusively determined (4, 62).

Defective axonal transport

A common theme in the neuropathology of PD that hasemerged over the last few years is the prominence of

DMV IN PD 7

Page 8: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

pathology in neurites. The earliest dopaminergic pathology inPD is thought to be a loss of axons projecting to the posteriorputamen (21, 64, 157, 244). Similar pathology to dopamineterminal fields in the striatum has been described in animalmodels of PD, including MPTP treatment in primates andmice and rotenone intoxication in rats, and the damage isapparent before loss of dopamine cell bodies in the substantianigra (42, 58, 129). Axonal damage has also been describedin other sites of PD pathology, such as the hippocampus,

where it is thought to correlate with cognitive dysfunction(23, 97). This has led to a theory suggesting that PD neuro-pathology may be a dying-back phenomenon progressingfrom loss of synaptic function to distal axonopathy andeventually to neuronal death (Fig. 5) (68, 110, 197, 286).

This has also been a burgeoning theme in the study of theautonomic nervous system in PD. AS neuritic pathology isoften the most prominent, only, or perhaps the earliest pa-thology observable in areas relevant to GI motility (6, 34,276). As discussed above, the DMV is not only a prominentsite of Lewy pathology in PD but also one of the mostcommon brain areas to be affected by incidental Lewy pa-thology, which is typically manifest as neuritic AS aggre-gation (35, 278).

Notwithstanding the general belief that axonal damage isthe earliest manifestation of PD pathology, there have beenfew direct investigations into the involvement of axon ter-minals in PD symptomatology and pathogenesis and none innoncatecholaminergic neurons, such as those in the DMV.However, investigations of the cardiac peripheral sympa-thetic nervous system in PD provide some interesting paral-lels. Sympathetic terminal loss is the neuropathologicalcorrelate to cardiovascular symptoms associated with par-kinsonism (110, 111). In the heart, cross-sectional postmor-tem studies suggest that damage to cardiac sympathetic nerveterminals precedes axon loss in the sympathetic nervoussystem and functions as a herald of neuron cell body pa-thology and loss (77, 93, 210, 211). DMV axons are very longand lightly myelinated meaning they may be particularlyvulnerable to axon-mediated injury, since their terminals arefar from the cell body at the end of a poorly protected andmetabolically demanding axon.

Mechanisms of nerve terminal and axon damage in PDhave not been fully elucidated (Fig. 6). Dopaminergic tox-ins like MPTP are concentrated in terminals by uptake viathe dopamine transporter whereupon they inhibit mito-chondria causing degeneration; this mechanism is active to alesser extent in norepinephrine neurons, like those in thepontine locus ceruleus (141, 203). Abnormal catecholamine

FIG. 5. PD as a dying-back neuropathy. This is a sche-matic hypothesis of a-synuclein aggregation and neuronalloss in the DMV in PD that begins in nerve terminals andaxons. As disease progresses, terminals and axons degeneratecausing symptoms. The process ultimately causes cell bodyloss in the DMV. Adapted from Orimo et al. (211).

FIG. 6. Nerve terminal degeneration in PD. This is a schematic hypothesis concerning possible mechanisms of DMVterminal damage in PD. There is a complex inter-relationship between a-synuclein, axon transport abnormalities, mito-chondrial dysfunction, inflammation, and oxidative stress. AS, a-synuclein; RNS, reactive nitrogen species; ROS, reactiveoxygen species.

8 GREENE

Page 9: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

metabolism, such as that seen in mice with genetic hy-poactivity of the vesicular monoamine transporter 2, has alsobeen shown to preferentially damage catecholaminergicnerve terminals before causing selective degeneration ofcatecholaminergic cell bodies (44, 264). MPTP disrupts ax-onal transport, which can lead to terminal degeneration byinterfering with the provision of necessary cellular compo-nents, such as mitochondria (29, 31, 68, 109, 197). As aresult, mitochondria in axons and terminals are under par-ticular stress, and studies demonstrate that synaptic mito-chondria are highly vulnerable to mitochondrial complex Iinhibition (65). Overexpression of AS has recently beenshown to disrupt axonal transport and cause morphologi-cal changes in dopamine terminals before cell body loss incellular models and in mice (56, 166). When viewed inthe context of the central role that mitochondria may playin PD pathogenesis, current results suggest that the com-bination of disrupted axonal transport and mitochondrialdysfunction may be particularly damaging (27, 47, 68, 215,218, 272, 290).

High metabolic demand and oxidative stress

High cellular energy demand has been described as a ro-bust marker for selectively vulnerable neurons in a variety ofneurodegenerative diseases (57, 113, 114). In PD, neuronsvulnerable to parkinsonian neuropathology, including thosein the DMV, have a high rate of metabolic demand. Onefactor in that demand is axonal transport through long, thin,highly branched, poorly myelinated axons as discussed above(33, 169), but synaptic activity is the main driver of theneuronal metabolic demand and consumes the majority ofmitochondrial energy produced (11, 146). Axon morphologyplays a role in this demand as well, since neurons of thisshape tend to be highly metabolically active, at least partiallyas a result of the greater energy requirement for neurotrans-mission in such cells. Transmission is inherently slower insmall-caliber fibers and poor myelination means there islimited energy benefit from saltatory conduction. Ad-ditionally, the energy cost of terminal maintenance is highdue their long axon length and resultant axonal transportdemands.

Adding to these factors is the recent discovery that DMVneurons, in addition to other neurons susceptible to PD, arevulnerable to metabolic stress due to their intrinsic activitypatterns and the mechanisms by which those patterns aregenerated and maintained (108). Cholinergic DMV neuronsexhibit an autonomous pacemaker activity accompanied by arobust extracellular calcium entry that is only weakly buff-ered by endogenous proteins. Neuronal activity and calciumentry both factor into elevating mitochondrial oxidant stressin DMV neurons. These characteristics are not unique toDMV neurons, but present in several neuronal subtypesvulnerable to degeneration in PD, including dopamine neu-rons in the substantia nigra and noradrenergic neurons in thelocus ceruleus, thereby suggesting a common phenotype thatdefines neurons at risk in PD (47, 48, 261).

This hypothesis is yet to be fully tested in vivo, but oxi-dative metabolic stress related to mitochondrial dysfunctionhas been hypothesized to be intimately involved in PDpathogenesis (272, 290). A deficiency in complex I of theelectron transport chain has been consistently described in

PD, and the complex I inhibitors, MPTP and rotenone, mimicbehavioral and neuropathological features of PD in animalmodels (25, 27, 64, 215, 272, 290). As might be expected,cells more dependent on mitochondrial respiration are moresusceptible to mitochondrial inhibitors such as rotenone. Theearliest behavioral abnormality associated with systemicadministration of rotenone to rats is delayed gastric empty-ing, a result suggesting that neurons regulating GI motility,such as those in the DMV, may be particularly vulnerable tomitochondrial inhibition as is suggested by the recent phys-iological data (115).

Synthesis and Future Directions

The DMV is uniquely vulnerable to damage from PD. Thisselective vulnerability appears to be based both on its anat-omy and intrinsic properties of its neurons. From an ana-tomical standpoint, efferent fibers in the vagus nerve from theDMV course throughout the GI tract forming a close linkbetween the peripheral and CNS and a point of proximalcontact between the environment (in the GI tract lumen) andbrainstem areas where PD pathology is believed to be set inmotion. In addition, since the GI tract is the largest immuneorgan in the body, vagal terminals are particularly exposed todamaging inflammatory insults. Intrinsically, DMV neuronsare under high levels of oxidative stress due to their expres-sion level of AS, fragile axons, and specific neuronal physi-ology. Moreover, several consequences of DMV damage,namely, GI dysfunction and unrestrained inflammation, maybe the contributing causes of neurodegeneration and propa-gate a vicious cycle of injury that consumes the DMV andspreads to other vulnerable brain regions.

Although perhaps two among many, the DMV and vagusnerve may be crucial nodes in the network of pathophysi-ology that ultimately leads to PD. Their accessible anatomymay prove beneficial for PD therapeutics in the future if itcan be harnessed, via neural stimulation, gene therapy, ortargeted pharmacological intervention, to provide a systemin which to study PD pathogenesis and a highway with di-rect access to neurons particularly vulnerable to the diseaseprocess.

Author Disclosure Statement

Dr. Greene has no conflicts of interest.

References

1. Abbott RA, Cox M, Markus H, and Tomkins A. Diet,body size and micronutrient status in Parkinson’s disease.Eur J Clin Nutr 46: 879–884, 1992.

2. Abbott RD, Petrovitch H, White LR, Masaki KH, TannerCM, Curb JD, Grandinetti A, Blanchette PL, Popper JS,and Ross GW. Frequency of bowel movements and thefuture risk of Parkinson’s disease. Neurology 57: 456–462, 2001.

3. Alvarez-Erviti L, Couch Y, Richardson J, Cooper JM, andWood MJ. Alpha-synuclein release by neurons activatesthe inflammatory response in a microglial cell line. Neu-rosci Res 69: 337–342, 2011.

4. Alvarez-Erviti L, Seow Y, Schapira AH, Gardiner C,Sargent IL, Wood MJ, and Cooper JM. Lysosomal dys-function increases exosome-mediated alpha-synuclein re-lease and transmission. Neurobiol Dis 42: 360–367, 2011.

DMV IN PD 9

Page 10: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

5. Angyan L. Medullary-induced alterations in intracranialself-stimulation from the substantia nigra. Neurobiology(Bp) 2: 195–210, 1994.

6. Annerino DM, Arshad S, Taylor GM, Adler CH, BeachTG, and Greene JG. Parkinson’s disease is not associatedwith gastrointestinal myenteric ganglion neuron loss. ActaNeuropathol 124: 665–680, 2012.

7. Arrasate M, Mitra S, Schweitzer ES, Segal MR, andFinkbeiner S. Inclusion body formation reduces levels ofmutant huntingtin and the risk of neuronal death. Nature431: 805–810, 2004.

8. Ashraf W, Pfeiffer R, Park F, Lof J, and Quigley EM.Constipation in Parkinson’s Disease: objective assessmentand response to psyllium. Mov Disord 12: 946–951, 1997.

9. Ashraf W, Wszolek ZK, Pfeiffer RF, Normand M, MaurerK, Srb F, Edwards LL, and Quigley EM. Anorectalfunction in fluctuating (on-off) Parkinson’s disease:evaluation by combined anorectal manometry and elec-tromyography. Mov Disord 10: 650–657, 1995.

10. Astarloa R, Mena M, Sanchez V, de la Vega L, and deYebenes J. Clinical and pharmacokinetic effects of a dietrich in insoluble fiber on Parkinson disease. Clin Neuro-pharmacol 15: 375–380, 1992.

11. Astrup J, Sorensen PM, and Sorensen HR. Oxygen andglucose consumption related to Na + -K + transport incanine brain. Stroke 12: 726–730, 1981.

12. Aziz NA, van der Marck MA, Pijl H, Olde Rikkert MG,Bloem BR, and Roos RA. Weight loss in neurodegener-ative disorders. J Neurol 255: 1872–1880, 2008.

13. Banati RB, Daniel SE, and Blunt SB. Glial pathology butabsence of apoptotic nigral neurons in long-standingParkinson’s disease. Mov Disord 13: 221–227, 1998.

14. Barcia C, Sanchez Bahillo A, Fernandez-Villalba E,Bautista V, Poza YPM, Fernandez-Barreiro A, Hirsch EC,and Herrero MT. Evidence of active microglia in sub-stantia nigra pars compacta of parkinsonian monkeys 1year after MPTP exposure. Glia 46: 402–409, 2004.

15. Barrett JC, Hansoul S, Nicolae DL, Cho JH, Duerr RH,Rioux JD, Brant SR, Silverberg MS, Taylor KD, BarmadaMM, Bitton A, Dassopoulos T, Datta LW, Green T,Griffiths AM, Kistner EO, Murtha MT, Regueiro MD,Rotter JI, Schumm LP, Steinhart AH, Targan SR, XavierRJ, Libioulle C, Sandor C, Lathrop M, Belaiche J, DewitO, Gut I, Heath S, Laukens D, Mni M, Rutgeerts P, VanGossum A, Zelenika D, Franchimont D, Hugot JP, de VosM, Vermeire S, Louis E, Cardon LR, Anderson CA,Drummond H, Nimmo E, Ahmad T, Prescott NJ, OnnieCM, Fisher SA, Marchini J, Ghori J, Bumpstead S,Gwilliam R, Tremelling M, Deloukas P, Mansfield J,Jewell D, Satsangi J, Mathew CG, Parkes M, Georges M,and Daly MJ. Genome-wide association defines more than30 distinct susceptibility loci for Crohn’s disease. NatGenet 40: 955–962, 2008.

16. Beach TG, Adler CH, Lue L, Sue LI, Bachalakuri J,Henry-Watson J, Sasse J, Boyer S, Shirohi S, Brooks R,Eschbacher J, White CL, 3rd, Akiyama H, Caviness J,Shill HA, Connor DJ, Sabbagh MN, and Walker DG.Unified staging system for Lewy body disorders: corre-lation with nigrostriatal degeneration, cognitive impair-ment and motor dysfunction. Acta Neuropathol 117: 613–634, 2009.

17. Beach TG, Adler CH, Sue LI, Vedders L, Lue L, White IiiCL, Akiyama H, Caviness JN, Shill HA, Sabbagh MN,and Walker DG. Multi-organ distribution of phosphory-

lated alpha-synuclein histopathology in subjects withLewy body disorders. Acta Neuropathol 119: 689–702,2010.

18. Benarroch EE. The central autonomic network: functionalorganization, dysfunction, and perspective. Mayo ClinProc 68: 988–1001, 1993.

19. Benarroch EE, Schmeichel AM, and Parisi JE. Involve-ment of the ventrolateral medulla in parkinsonism withautonomic failure. Neurology 54: 963–968, 2000.

20. Benner EJ, Banerjee R, Reynolds AD, Sherman S, PisarevVM, Tsiperson V, Nemachek C, Ciborowski P, Przed-borski S, Mosley RL, and Gendelman HE. Nitrated alpha-synuclein immunity accelerates degeneration of nigraldopaminergic neurons. PLoS One 3: e1376, 2008.

21. Bernheimer H, Birkmayer W, Hornykiewicz O, JellingerK, and Seitelberger F. Brain dopamine and the syndromesof Parkinson and Huntington. Clinical, morphological andneurochemical correlations. J Neurol Sci 20: 415–455,1973.

22. Berthoud HR, Carlson NR, and Powley TL. Topographyof efferent vagal innervation of the rat gastrointestinaltract. Am J Physiol 260: R200–R207, 1991.

23. Bertrand E, Lechowicz W, Szpak GM, Lewandowska E,Dymecki J, and Wierzba-Bobrowicz T. Limbic neuropa-thology in idiopathic Parkinson’s disease with concomi-tant dementia. Folia Neuropathol 42: 141–150, 2004.

24. Betarbet R, Sherer TB, and Greenamyre JT. Ubiquitin-proteasome system and Parkinson’s diseases. Exp Neurol191 Suppl 1: S17–S27, 2005.

25. Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M,Panov AV, and Greenamyre JT. Chronic systemic pesti-cide exposure reproduces features of Parkinson’s disease.Nat Neurosci 3: 1301–1306, 2000.

26. Bialecka M, Kurzawski M, Klodowska-Duda G, Opala G,Juzwiak S, Kurzawski G, Tan EK, and Drozdzik M.CARD15 variants in patients with sporadic Parkinson’sdisease. Neurosci Res 57: 473–476, 2007.

27. Bindoff LA, Birch-Machin M, Cartlidge NE, Parker WD,Jr., and Turnbull DM. Mitochondrial function in Parkin-son’s disease. Lancet 2: 49, 1989.

28. Bjarnason IT, Charlett A, Dobbs RJ, Dobbs SM, IbrahimMA, Kerwin RW, Mahler RF, Oxlade NL, Peterson DW,Plant JM, Price AB, and Weller C. Role of chronic in-fection and inflammation in the gastrointestinal tract in theetiology and pathogenesis of idiopathic parkinsonism. Part2: response of facets of clinical idiopathic parkinsonism toHelicobacter pylori eradication. A randomized, double-blind, placebo-controlled efficacy study. Helicobacter 10:276–287, 2005.

29. Boldogh IR and Pon LA. Mitochondria on the move.Trends Cell Biol 17: 502–510, 2007.

30. Bordia T, Parameswaran N, Fan H, Langston JW, McIn-tosh JM, and Quik M. Partial recovery of striatal nicotinicreceptors in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine(MPTP)-lesioned monkeys with chronic oral nicotine. JPharmacol Exp Ther 319: 285–292, 2006.

31. Borland MK, Trimmer PA, Rubinstein JD, Keeney PM,Mohanakumar K, Liu L, and Bennett JP, Jr. Chronic, low-dose rotenone reproduces Lewy neurites found in earlystages of Parkinson’s disease, reduces mitochondrialmovement and slowly kills differentiated SH-SY5Y neu-ral cells. Mol Neurodegener 3: 21, 2008.

32. Borovikova LV, Ivanova S, Zhang M, Yang H, BotchkinaGI, Watkins LR, Wang H, Abumrad N, Eaton JW, and

10 GREENE

Page 11: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

Tracey KJ. Vagus nerve stimulation attenuates the sys-temic inflammatory response to endotoxin. Nature 405:458–462, 2000.

33. Braak H, Bohl JR, Muller CM, Rub U, de Vos RA, andDel Tredici K. Stanley Fahn Lecture 2005: the stagingprocedure for the inclusion body pathology associatedwith sporadic Parkinson’s disease reconsidered. MovDisord 21: 2042–2051, 2006.

34. Braak H, de Vos RA, Bohl J, and Del Tredici K. Gastricalpha-synuclein immunoreactive inclusions in Meissner’sand Auerbach’s plexuses in cases staged for Parkinson’sdisease-related brain pathology. Neurosci Lett 396: 67–72,2006.

35. Braak H, Del Tredici K, Rub U, de Vos RA, Jansen SteurEN, and Braak E. Staging of brain pathology related tosporadic Parkinson’s disease. Neurobiol Aging 24: 197–211, 2003.

36. Braak H, Rub U, Gai WP, and Del Tredici K. IdiopathicParkinson’s disease: possible routes by which vulnerableneuronal types may be subject to neuroinvasion by anunknown pathogen. J Neural Transm 110: 517–536, 2003.

37. Brochard V, Combadiere B, Prigent A, Laouar Y, PerrinA, Beray-Berthat V, Bonduelle O, Alvarez-Fischer D,Callebert J, Launay JM, Duyckaerts C, Flavell RA, HirschEC, and Hunot S. Infiltration of CD4 + lymphocytes intothe brain contributes to neurodegeneration in a mousemodel of Parkinson disease. J Clin Invest 119: 182–192,2009.

38. Bronstein DM, Perez-Otano I, Sun V, Mullis Sawin SB,Chan J, Wu GC, Hudson PM, Kong LY, Hong JS, andMcMillian MK. Glia-dependent neurotoxicity and neuro-protection in mesencephalic cultures. Brain Res 704: 112–116, 1995.

39. Brown P. Abnormal oscillatory synchronisation in themotor system leads to impaired movement. Curr OpinNeurobiol 17: 656–664, 2007.

40. Brown P, Oliviero A, Mazzone P, Insola A, Tonali P, andDi Lazzaro V. Dopamine dependency of oscillations be-tween subthalamic nucleus and pallidum in Parkinson’sdisease. J Neurosci 21: 1033–1038, 2001.

41. Burke RE, Dauer WT, and Vonsattel JP. A critical eval-uation of the Braak staging scheme for Parkinson’s dis-ease. Ann Neurol 64: 485–491, 2008.

42. Burns RS, Chiueh CC, Markey SP, Ebert MH, JacobowitzDM, and Kopin IJ. A primate model of parkinsonism:selective destruction of dopaminergic neurons in the parscompacta of the substantia nigra by N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Proc Natl Acad Sci U S A 80:4546–4550, 1983.

43. Cai QQ, Zheng LF, Fan RF, Lian H, Zhou L, Song HY,Tang YY, Feng XY, Guo ZK, Wang ZY, and Zhu JX.Distribution of dopamine receptors D1- and D2-immu-noreactive neurons in the dorsal motor nucleus of vagus inrats. Auton Neurosci 176: 48–53, 2013.

44. Caudle WM, Richardson JR, Wang MZ, Taylor TN,Guillot TS, McCormack AL, Colebrooke RE, Di MonteDA, Emson PC, and Miller GW. Reduced vesicular stor-age of dopamine causes progressive nigrostriatal neuro-degeneration. J Neurosci 27: 8138–8148, 2007.

45. Cechetto DF. Central representation of visceral function.Fed Proc 46: 17–23, 1987.

46. Cechetto DF and Saper CB. Evidence for a viscerotopicsensory representation in the cortex and thalamus in therat. J Comp Neurol 262: 27–45, 1987.

47. Chan CS, Gertler TS, and Surmeier DJ. Calcium homeo-stasis, selective vulnerability and Parkinson’s disease.Trends Neurosci 32: 249–256, 2009.

48. Chan CS, Guzman JN, Ilijic E, Mercer JN, Rick C, TkatchT, Meredith GE, and Surmeier DJ. ‘Rejuvenation’ protectsneurons in mouse models of Parkinson’s disease. Nature447: 1081–1086, 2007.

49. Charles PD, Van Blercom N, Krack P, Lee SL, Xie J,Besson G, Benabid AL, and Pollak P. Predictors of ef-fective bilateral subthalamic nucleus stimulation for PD.Neurology 59: 932–934, 2002.

50. Chen CL, Lin HH, Chen SY, and Lin SZ. Utility ofelectrogastrography in differentiating Parkinson’s diseasewith or without gastrointestinal symptoms: a prospectivecontrolled study. Digestion 71: 187–191, 2005.

51. Chen H, Jacobs E, Schwarzschild MA, McCullough ML,Calle EE, Thun MJ, and Ascherio A. Nonsteroidal anti-inflammatory drug use and the risk for Parkinson’s dis-ease. Ann Neurol 58: 963–967, 2005.

52. Chen H, Zhang SM, Hernan MA, Schwarzschild MA,Willett WC, Colditz GA, Speizer FE, and Ascherio A.Nonsteroidal anti-inflammatory drugs and the risk ofParkinson disease. Arch Neurol 60: 1059–1064, 2003.

53. Cheyuo C, Jacob A, Wu R, Zhou M, Coppa GF, and WangP. The parasympathetic nervous system in the quest forstroke therapeutics. J Cereb Blood Flow Metab 31: 1187–1195, 2011.

54. Chiba T, Kayahara T, and Nakano K. Efferent projectionsof infralimbic and prelimbic areas of the medial prefrontalcortex in the Japanese monkey, Macaca fuscata. Brain Res888: 83–101, 2001.

55. This reference has been deleted.56. Chung CY, Koprich JB, Siddiqi H, and Isacson O. Dy-

namic changes in presynaptic and axonal transport pro-teins combined with striatal neuroinflammation precededopaminergic neuronal loss in a rat model of AAV alpha-synucleinopathy. J Neurosci 29: 3365–3373, 2009.

57. Chung CY, Seo H, Sonntag KC, Brooks A, Lin L, andIsacson O. Cell type-specific gene expression of midbraindopaminergic neurons reveals molecules involved in theirvulnerability and protection. Hum Mol Genet 14: 1709–1725, 2005.

58. Cochiolo JA, Ehsanian R, and Bruck DK. Acute ultra-structural effects of MPTP on the nigrostriatal pathway ofthe C57BL/6 adult mouse: evidence of compensatoryplasticity in nigrostriatal neurons. J Neurosci Res 59: 126–135, 2000.

59. Connor DE, Jr., Nixon M, Nanda A, and Guthikonda B.Vagal nerve stimulation for the treatment of medicallyrefractory epilepsy: a review of the current literature.Neurosurg Focus 32: E12, 2012.

60. Cookson MR and van der Brug M. Cell systems and thetoxic mechanism(s) of alpha-synuclein. Exp Neurol 209:5–11, 2008.

61. Costa RM, Lin SC, Sotnikova TD, Cyr M, GainetdinovRR, Caron MG, and Nicolelis MA. Rapid alterations incorticostriatal ensemble coordination during acute dopa-mine-dependent motor dysfunction. Neuron 52: 359–369,2006.

62. Danzer KM, Ruf WP, Putcha P, Joyner D, Hashimoto T,Glabe C, Hyman BT, and McLean PJ. Heat-shock protein70 modulates toxic extracellular alpha-synuclein oligo-mers and rescues trans-synaptic toxicity. FASEB J 25:326–336, 2011.

DMV IN PD 11

Page 12: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

63. Darbin O and Wichmann T. Effects of striatal GABA A-receptor blockade on striatal and cortical activity inmonkeys. J Neurophysiol 99: 1294–1305, 2008.

64. Dauer W and Przedborski S. Parkinson’s disease: mech-anisms and models. Neuron 39: 889–909, 2003.

65. Davey GP, Peuchen S, and Clark JB. Energy thresholds inbrain mitochondria. Potential involvement in neurode-generation. J Biol Chem 273: 12753–12757, 1998.

66. Davies KN, King D, Billington D, and Barrett JA. In-testinal permeability and orocaecal transit time in elderlypatients with Parkinson’s disease. Postgrad Med J 72:164–167, 1996.

67. de Jong W, Linthorst AC, and Versteeg HG. The nigros-triatal dopamine system and the development of hyper-tension in the spontaneously hypertensive rat. Arch MalCoeur Vaiss 88: 1193–1196, 1995.

68. De Vos KJ, Grierson AJ, Ackerley S, and Miller CC. Roleof axonal transport in neurodegenerative diseases. AnnuRev Neurosci 31: 151–173, 2008.

69. Dejean C, Gross CE, Bioulac B, and Boraud T. Dynamicchanges in the cortex-basal ganglia network after dopaminedepletion in the rat. J Neurophysiol 100: 385–396, 2008.

70. Dejean C, Hyland B, and Arbuthnott G. Cortical effects ofsubthalamic stimulation correlate with behavioral recov-ery from dopamine antagonist induced akinesia. CerebCortex 19: 1055–1063, 2009.

71. Delong M and Juncos J. Parkinson’s disease and othermovement disorders. In: Harrison’s Principles of InternalMedicine, edited by Kasper DL. New York: McGraw-HillMedical, 2004, pp. 2406–2418.

72. Derkinderen P, Rouaud T, Lebouvier T, Bruley des Var-annes S, Neunlist M, De and Giorgio R. Parkinson dis-ease: the enteric nervous system spills its guts. Neurology77: 1761–1767, 2011.

73. Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E,Crews L, Spencer B, Masliah E, and Lee SJ. Inclusionformation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl AcadSci U S A 106: 13010–13015, 2009.

74. Devi L, Raghavendran V, Prabhu BM, Avadhani NG, andAnandatheerthavarada HK. Mitochondrial import andaccumulation of alpha-synuclein impair complex I in hu-man dopaminergic neuronal cultures and Parkinson dis-ease brain. J Biol Chem 283: 9089–9100, 2008.

75. Devos D, Lebouvier T, Lardeux B, Biraud M, Rouaud T,Pouclet H, Coron E, Bruley des Varannes S, Naveilhan P,Nguyen JM, Neunlist M, and Derkinderen P. Colonic in-flammation in Parkinson’s disease. Neurobiol Dis 50: 42–48, 2013.

76. Diamond B and Tracey KJ. Mapping the immunologicalhomunculus. Proc Natl Acad Sci U S A 108: 3461–3462,2011.

77. Dickson DW, Fujishiro H, DelleDonne A, Menke J,Ahmed Z, Klos KJ, Josephs KA, Frigerio R, Burnett M,Parisi JE, and Ahlskog JE. Evidence that incidental Lewybody disease is pre-symptomatic Parkinson’s disease. ActaNeuropathol 115: 437–444, 2008.

78. Djaldetti R, Achiron A, Ziv I, and Melamed E. Firstemergence of ‘‘delayed-on’’ and ‘‘dose failure’’ phe-nomena in a patient with Parkinson’s disease followingvagotomy. Mov Disord 9: 582–583, 1994.

79. Djaldetti R, Baron J, Ziv I, and Melamed E. Gastricemptying in Parkinson’s disease: patients with and withoutresponse fluctuations. Neurology 46: 1051–1054, 1996.

80. Djaldetti R, Ziv I, and Melamed E. Impaired absorption oforal levodopa: a major cause for response fluctuations inParkinson’s disease. Isr J Med Sci 32: 1224–1227, 1996.

81. Dobbs RJ, Dobbs SM, Weller C, Bjarnason IT, OxladeNL, Charlett A, Al-Janabi MA, Kerwin RW, Mahler RF,and Price AB. Role of chronic infection and inflammationin the gastrointestinal tract in the etiology and pathogen-esis of idiopathic parkinsonism. Part 1: eradication ofHelicobacter in the cachexia of idiopathic parkinsonism.Helicobacter 10: 267–275, 2005.

82. Eadie MJ. The pathology of certain medullary nuclei inparkinsonism. Brain 86: 781–792, 1963.

83. Edwards LL, Pfeiffer RF, Quigley EM, Hofman R, andBalluff M. Gastrointestinal symptoms in Parkinson’s dis-ease. Mov Disord 6: 151–156, 1991.

84. Edwards LL, Quigley EM, Harned RK, Hofman R, andPfeiffer RF. Characterization of swallowing and defeca-tion in Parkinson’s disease. Am J Gastroenterol 89: 15–25, 1994.

85. Edwards LL, Quigley EM, and Pfeiffer RF. Gastro-intestinal dysfunction in Parkinson’s disease: frequencyand pathophysiology. Neurology 42: 726–732, 1992.

86. Fearnley JM and Lees AJ. Ageing and Parkinson’s dis-ease: substantia nigra regional selectivity. Brain 114 (Pt5): 2283–2301, 1991.

87. Filion M. Effects of interruption of the nigrostriatalpathway and of dopaminergic agents on the spontaneousactivity of globus pallidus neurons in the awake monkey.Brain Res 178: 425–441, 1979.

88. Foix C and Nicolesco J. Anatomie Cerebrale: Les NoyauxGris Centraux et la Region Mesencephalo-soux-optique.Pairs: Masson, 1925.

89. Forsyth CB, Shannon KM, Kordower JH, Voigt RM,Shaikh M, Jaglin JA, Estes JD, Dodiya HB, and Ke-shavarzian A. Increased intestinal permeability correlateswith sigmoid mucosa alpha-synuclein staining and endo-toxin exposure markers in early Parkinson’s disease. PLoSOne 6: e28032, 2011.

90. Francois C, Tande D, Yelnik J, and Hirsch EC. Distribu-tion and morphology of nigral axons projecting to thethalamus in primates. J Comp Neurol 447: 249–260, 2002.

91. Frank-Cannon TC, Tran T, Ruhn KA, Martinez TN, HongJ, Marvin M, Hartley M, Trevino I, O’Brien DE, Casey B,Goldberg MS, and Tansey MG. Parkin deficiency in-creases vulnerability to inflammation-related nigral de-generation. J Neurosci 28: 10825–10834, 2008.

92. Fudge JL, Breitbart MA, Danish M, and Pannoni V. In-sular and gustatory inputs to the caudal ventral striatum inprimates. J Comp Neurol 490: 101–118, 2005.

93. Fujishiro H, Frigerio R, Burnett M, Klos KJ, Josephs KA,Delledonne A, Parisi JE, Ahlskog JE, and Dickson DW.Cardiac sympathetic denervation correlates with clinicaland pathologic stages of Parkinson’s disease. Mov Disord23: 1085–1092, 2008.

94. Gai WP, Blessing WW, and Blumbergs PC. Ubiquitin-positive degenerating neurites in the brainstem in Par-kinson’s disease. Brain 118 (Pt 6): 1447–1459, 1995.

95. Gai WP, Blumbergs PC, Geffen LB, and Blessing WW.Age-related loss of dorsal vagal neurons in Parkinson’sdisease. Neurology 42: 2106–2111, 1992.

96. Gai WP, Geffen LB, Denoroy L, and Blessing WW. Lossof C1 and C3 epinephrine-synthesizing neurons in themedulla oblongata in Parkinson’s disease. Ann Neurol 33:357–367, 1993.

12 GREENE

Page 13: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

97. Galvin JE, Uryu K, Lee VM, and Trojanowski JQ. Axonpathology in Parkinson’s disease and Lewy body dementiahippocampus contains alpha-, beta-, and gamma-synuclein.Proc Natl Acad Sci U S A 96: 13450–13455, 1999.

98. Gao HM, Kotzbauer PT, Uryu K, Leight S, TrojanowskiJQ, and Lee VM. Neuroinflammation and oxidation/ni-tration of alpha-synuclein linked to dopaminergic neuro-degeneration. J Neurosci 28: 7687–7698, 2008.

99. Gao HM, Liu B, Zhang W, and Hong JS. Synergisticdopaminergic neurotoxicity of MPTP and inflammogenlipopolysaccharide: relevance to the etiology of Parkin-son’s disease. FASEB J 17: 1957–1959, 2003.

100. Gao HM, Zhang F, Zhou H, Kam W, Wilson B, and HongJS. Neuroinflammation and alpha-synuclein dysfunctionpotentiate each other, driving chronic progression ofneurodegeneration in a mouse model of Parkinson’s dis-ease. Environ Health Perspect 119: 807–814, 2011.

101. Gatev P, Darbin O, and Wichmann T. Oscillations in thebasal ganglia under normal conditions and in movementdisorders. Mov Disord 21: 1566–1577, 2006.

102. Gatev P and Wichmann T. Interactions between corticalrhythms and spiking activity of single basal ganglia neu-rons in the normal and parkinsonian state. Cereb Cortex19: 1330–1344, 2009.

103. German DC, Manaye KF, Sonsalla PK, and Brooks BA.Midbrain dopaminergic cell loss in Parkinson’s diseaseand MPTP-induced parkinsonism: sparing of calbindin-D28k-containing cells. Ann N Y Acad Sci 648: 42–62,1992.

104. German DC, Manaye KF, White CL, 3rd, Woodward DJ,McIntire DD, Smith WK, Kalaria RN, and Mann DM.Disease-specific patterns of locus coeruleus cell loss. AnnNeurol 32: 667–676, 1992.

105. Ghashghaei HT and Barbas H. Neural interaction betweenthe basal forebrain and functionally distinct prefrontalcortices in the rhesus monkey. Neuroscience 103: 593–614, 2001.

106. Goetze O, Nikodem AB, Wiezcorek J, Banasch M,Przuntek H, Mueller T, Schmidt WE, and Woitalla D.Predictors of gastric emptying in Parkinson’s disease.Neurogastroenterol Motil 18: 369–375, 2006.

107. Goetze O, Wieczorek J, Mueller T, Przuntek H, SchmidtWE, and Woitalla D. Impaired gastric emptying of a solidtest meal in patients with Parkinson’s disease using 13C-sodium octanoate breath test. Neurosci Lett 375: 170–173,2005.

108. Goldberg JA, Guzman JN, Estep CM, Ilijic E, KondapalliJ, Sanchez-Padilla J, and Surmeier DJ. Calcium entry in-duces mitochondrial oxidant stress in vagal neurons atrisk in Parkinson’s disease. Nat Neurosci 15: 1414–1421,2012.

109. Goldstein AY, Wang X, and Schwarz TL. Axonal trans-port and the delivery of pre-synaptic components. CurrOpin Neurobiol 18: 495–503, 2008.

110. Goldstein DS. Cardiac denervation in patients with Par-kinson disease. Cleve Clin J Med 74 Suppl 1: S91–S94,2007.

111. Goldstein DS, Sharabi Y, Karp BI, Bentho O, Saleem A,Pacak K, and Eisenhofer G. Cardiac sympathetic dener-vation preceding motor signs in Parkinson disease. ClinAuton Res 17: 118–121, 2007.

112. Gravante G, Sabatino M, Caravaglios G, and La Grutta V.Nigral influence on intestinal activity in the cat. ActaNeurol (Napoli) 10: 231–238, 1988.

113. Greene JG. Gene expression profiles of brain dopamineneurons and relevance to neuropsychiatric disease. JPhysiol 575: 411–416, 2006.

114. Greene JG, Dingledine R, and Greenamyre JT. Gene ex-pression profiling of rat midbrain dopamine neurons: im-plications for selective vulnerability in parkinsonism.Neurobiol Dis 18: 19–31, 2005.

115. Greene JG, Noorian AR, and Srinivasan S. Delayed gas-tric emptying and enteric nervous system dysfunction inthe rotenone model of Parkinson’s disease. Exp Neurol218: 154–161, 2009.

116. Guerci B, Bourgeois C, Bresler L, Scherrer ML, andBohme P. Gastric electrical stimulation for the treatment ofdiabetic gastroparesis. Diabetes Metab 38: 393–402, 2012.

117. Haack DG, Baumann RJ, McKean HE, Jameson HD, andTurbek JA. Nicotine exposure and Parkinson disease. AmJ Epidemiol 114: 191–200, 1981.

118. Haber SN, Kunishio K, Mizobuchi M, and Lynd-Balta E.The orbital and medial prefrontal circuit through the pri-mate basal ganglia. J Neurosci 15: 4851–4867, 1995.

119. Halliday GM, Blumbergs PC, Cotton RG, Blessing WW,and Geffen LB. Loss of brainstem serotonin- and sub-stance P-containing neurons in Parkinson’s disease. BrainRes 510: 104–107, 1990.

120. Halliday GM, Li YW, Blumbergs PC, Joh TH, Cotton RG,Howe PR, Blessing WW, and Geffen LB. Neuropathologyof immunohistochemically identified brainstem neurons inParkinson’s disease. Ann Neurol 27: 373–385, 1990.

121. Hammond C, Bergman H, and Brown P. Pathologicalsynchronization in Parkinson’s disease: networks, modelsand treatments. Trends Neurosci 30: 357–364, 2007.

122. Han YF and Cao GW. Role of nuclear receptor NR4A2 ingastrointestinal inflammation and cancers. World J Gas-troenterol 18: 6865–6873, 2012.

123. Hansen C, Angot E, Bergstrom AL, Steiner JA, Pieri L,Paul G, Outeiro TF, Melki R, Kallunki P, Fog K, Li JY,and Brundin P. alpha-Synuclein propagates from mousebrain to grafted dopaminergic neurons and seeds aggre-gation in cultured human cells. J Clin Invest 121: 715–725, 2011.

124. Hansen C and Li JY. Beyond alpha-synuclein transfer:pathology propagation in Parkinson’s disease. Trends MolMed 18: 248–255, 2012.

125. Hardoff R, Sula M, Tamir A, Soil A, Front A, Badarna S,Honigman S, and Giladi N. Gastric emptying time andgastric motility in patients with Parkinson’s disease. MovDisord 16: 1041–1047, 2001.

126. Hassler R. Zur Normalanatomie der Substantia nigra.Versuch einer architektonischen Gliederung. J PsychNeurol 48: 1–55, 1937.

127. Heimer G, Bar-Gad I, Goldberg JA, and Bergman H.Dopamine replacement therapy reverses abnormal syn-chronization of pallidal neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine primate model of parkinsonism.J Neurosci 22: 7850–7855, 2002.

128. Hely MA, Morris JG, Reid WG, and Trafficante R. Syd-ney Multicenter Study of Parkinson’s disease: non-L-dopa-responsive problems dominate at 15 years. MovDisord 20: 190–199, 2005.

129. Herkenham M, Little MD, Bankiewicz K, Yang SC,Markey SP, and Johannessen JN. Selective retention ofMPP + within the monoaminergic systems of the primatebrain following MPTP administration: an in vivo autora-diographic study. Neuroscience 40: 133–158, 1991.

DMV IN PD 13

Page 14: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

130. Hinault MP, Farina-Henriquez-Cuendet A, and Golou-binoff P. Molecular chaperones and associated cellularclearance mechanisms against toxic protein conformers inParkinson’s disease. Neurodegener Dis 8: 397–412, 2011.

131. Hirsch E, Graybiel AM, and Agid YA. Melanized dopa-minergic neurons are differentially susceptible to degener-ation in Parkinson’s disease. Nature 334: 345–348, 1988.

132. Hirsch EC and Hunot S. Neuroinflammation in Parkin-son’s disease: a target for neuroprotection? Lancet Neurol8: 382–397, 2009.

133. Hoeger S, Bergstraesser C, Selhorst J, Fontana J, Birck R,Waldherr R, Beck G, Sticht C, Seelen MA, van Son WJ,Leuvenink H, Ploeg R, Schnuelle P, and Yard BA.Modulation of brain dead induced inflammation by vagusnerve stimulation. Am J Transplant 10: 477–489, 2010.

134. Holmberg B, Corneliusson O, and Elam M. Bilateralstimulation of nucleus subthalamicus in advanced Par-kinson’s disease: no effects on, and of, autonomic dys-function. Mov Disord 20: 976–981, 2005.

135. Hsu LJ, Sagara Y, Arroyo A, Rockenstein E, Sisk A,Mallory M, Wong J, Takenouchi T, Hashimoto M, andMasliah E. alpha-synuclein promotes mitochondrial defi-cit and oxidative stress. Am J Pathol 157: 401–410, 2000.

136. Hu X, Zhang D, Pang H, Caudle WM, Li Y, Gao H, LiuY, Qian L, Wilson B, Di Monte DA, Ali SF, Zhang J,Block ML, and Hong JS. Macrophage antigen complex-1mediates reactive microgliosis and progressive dopami-nergic neurodegeneration in the MPTP model of Parkin-son’s disease. J Immunol 181: 7194–7204, 2008.

137. Huston JM, Ochani M, Rosas-Ballina M, Liao H, OchaniK, Pavlov VA, Gallowitsch-Puerta M, Ashok M, CzuraCJ, Foxwell B, Tracey KJ, and Ulloa L. Splenectomyinactivates the cholinergic antiinflammatory pathwayduring lethal endotoxemia and polymicrobial sepsis. J ExpMed 203: 1623–1628, 2006.

138. Ito S and Craig AD. Striatal projections of the vagal-responsive region of the thalamic parafascicular nucleusin macaque monkeys. J Comp Neurol 506: 301–327, 2008.

139. Iwanaga K, Wakabayashi K, Yoshimoto M, Tomita I,Satoh H, Takashima H, Satoh A, Seto M, Tsujihata M, andTakahashi H. Lewy body-type degeneration in cardiacplexus in Parkinson’s and incidental Lewy body diseases.Neurology 52: 1269–1271, 1999.

140. Jang H, Boltz D, Sturm-Ramirez K, Shepherd KR, Jiao Y,Webster R, and Smeyne RJ. Highly pathogenic H5N1influenza virus can enter the central nervous system andinduce neuroinflammation and neurodegeneration. ProcNatl Acad Sci U S A 106: 14063–14068, 2009.

141. Javitch JA, D’Amato RJ, Strittmatter SM, and Snyder SH.Parkinsonism-inducing neurotoxin, N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine: uptake of the metabolite N-methyl-4-phenylpyridine by dopamine neurons explainsselective toxicity. Proc Natl Acad Sci U S A 82: 2173–2177, 1985.

142. Johnson LR. Gastrointestinal Physiology. St. Louis:Mosby, Inc., 2001.

143. Jost WH and Schrank B. Defecatory disorders in de novoParkinsonians—colonic transit and electromyogram of theexternal anal sphincter. Wien Klin Wochenschr 110: 535–537, 1998.

144. Kalaitzakis ME, Graeber MB, Gentleman SM, and PearceRK. Controversies over the staging of alpha-synucleinpathology in Parkinson’s disease. Acta Neuropathol 116:125–128; author reply 129–131, 2008.

145. Kalaitzakis ME, Graeber MB, Gentleman SM, and PearceRK. The dorsal motor nucleus of the vagus is not anobligatory trigger site of Parkinson’s disease: a criticalanalysis of alpha-synuclein staging. Neuropathol ApplNeurobiol 34: 284–295, 2008.

146. Kann O and Kovacs R. Mitochondria and neuronal ac-tivity. Am J Physiol Cell Physiol 292: C641–C657, 2007.

147. Kashihara K. Weight loss in Parkinson’s disease. J Neurol253 Suppl 7: VII38–VII41, 2006.

148. Kawamata J, Suzuki S, and Shimohama S. Alpha7 nicotinicacetylcholine receptor mediated neuroprotection in Par-kinson’s disease. Curr Drug Targets 13: 623–630, 2012.

149. Kessler, II and Diamond EL. Epidemiologic studies of Par-kinson’s disease. I. Smoking and Parkinson’s disease: a surveyand explanatory hypothesis. Am J Epidemiol 94: 16–25, 1971.

150. Komisaruk BR, Whipple B, Crawford A, Liu WC, KalninA, and Mosier K. Brain activation during vaginocervicalself-stimulation and orgasm in women with completespinal cord injury: fMRI evidence of mediation by thevagus nerves. Brain Res 1024: 77–88, 2004.

151. Kordower JH, Chu Y, Hauser RA, Freeman TB, andOlanow CW. Lewy body-like pathology in long-termembryonic nigral transplants in Parkinson’s disease. NatMed 14: 504–506, 2008.

152. Kordower JH, Chu Y, Hauser RA, Olanow CW, andFreeman TB. Transplanted dopaminergic neurons developPD pathologic changes: a second case report. Mov Disord23: 2303–2306, 2008.

153. Krygowska-Wajs A, Cheshire WP, Jr., Wszolek ZK,Hubalewska-Dydejczyk A, Jasinska-Myga B, Farrer MJ,Moskala M, and Sowa-Staszczak A. Evaluation of gastricemptying in familial and sporadic Parkinson disease.Parkinsonism Relat Disord 15: 692–696, 2009.

154. Kumaria A and Tolias CM. Is there a role for vagus nervestimulation therapy as a treatment of traumatic brain in-jury? Br J Neurosurg 26: 316–320, 2012.

155. Kupsky WJ, Grimes MM, Sweeting J, Bertsch R, and CoteLJ. Parkinson’s disease and megacolon: concentric hya-line inclusions (Lewy bodies) in enteric ganglion cells.Neurology 37: 1253–1255, 1987.

156. Kurlan R, Rothfield KP, Woodward WR, Nutt JG, MillerC, Lichter D, and Shoulson I. Erratic gastric emptying oflevodopa may cause ‘‘random’’ fluctuations of parkinso-nian mobility. Neurology 38: 419–421, 1988.

157. Lach B, Grimes D, Benoit B, and Minkiewicz-Janda A.Caudate nucleus pathology in Parkinson’s disease: ultra-structural and biochemical findings in biopsy material.Acta Neuropathol 83: 352–360, 1992.

158. Ladabaum U, Minoshima S, Hasler WL, Cross D, CheyWD, and Owyang C. Gastric distention correlates withactivation of multiple cortical and subcortical regions.Gastroenterology 120: 369–376, 2001.

159. Langston JW. The Parkinson’s complex: parkinsonismis just the tip of the iceberg. Ann Neurol 59: 591–596,2006.

160. Langston JW, Forno LS, Tetrud J, Reeves AG, Kaplan JA,and Karluk D. Evidence of active nerve cell degenerationin the substantia nigra of humans years after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine exposure. Ann Neurol46: 598–605, 1999.

161. Lanotte M, Lopiano L, Torre E, Bergamasco B, Colloca L,and Benedetti F. Expectation enhances autonomic re-sponses to stimulation of the human subthalamic limbicregion. Brain Behav Immun 19: 500–509, 2005.

14 GREENE

Page 15: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

162. Le WD, Xu P, Jankovic J, Jiang H, Appel SH, Smith RG,and Vassilatis DK. Mutations in NR4A2 associated withfamilial Parkinson disease. Nat Genet 33: 85–89, 2003.

163. Lebouvier T, Chaumette T, Damier P, Coron E, Touche-feu Y, Vrignaud S, Naveilhan P, Galmiche JP, Bruley desVarannes S, Derkinderen P, and Neunlist M. Pathologicallesions in colonic biopsies during Parkinson’s disease. Gut57: 1741–1743, 2008.

164. Lebouvier T, Chaumette T, Paillusson S, Duyckaerts C,Bruley des Varannes S, Neunlist M, and Derkinderen P.The second brain and Parkinson’s disease. Eur J Neurosci30: 735–741, 2009.

165. Lebouvier T, Neunlist M, Bruley des Varannes S, CoronE, Drouard A, N’Guyen JM, Chaumette T, Tasselli M,Paillusson S, Flamand M, Galmiche JP, Damier P, andDerkinderen P. Colonic biopsies to assess the neuropa-thology of Parkinson’s disease and its relationship withsymptoms. PLoS One 5: e12728, 2010.

166. Lee HJ, Khoshaghideh F, Lee S, and Lee SJ. Impairmentof microtubule-dependent trafficking by overexpression ofalpha-synuclein. Eur J Neurosci 24: 3153–3162, 2006.

167. Lee HJ, Suk JE, Patrick C, Bae EJ, Cho JH, Rho S,Hwang D, Masliah E, and Lee SJ. Direct transfer ofalpha-synuclein from neuron to astroglia causes inflam-matory responses in synucleinopathies. J Biol Chem 285:9262–9272, 2010.

168. Lee JK, McCoy MK, Harms AS, Ruhn KA, Gold SJ, andTansey MG. Regulator of G-protein signaling 10 pro-motes dopaminergic neuron survival via regulation of themicroglial inflammatory response. J Neurosci 28: 8517–8528, 2008.

169. Lees AJ. The Parkinson chimera. Neurology 72: S2–S11,2009.

170. Levy R, Ashby P, Hutchison WD, Lang AE, Lozano AM,and Dostrovsky JO. Dependence of subthalamic nucleusoscillations on movement and dopamine in Parkinson’sdisease. Brain 125: 1196–1209, 2002.

171. Lewis SJ and Barker RA. Understanding the dopaminer-gic deficits in Parkinson’s disease: insights into diseaseheterogeneity. J Clin Neurosci 16: 620–625, 2009.

172. Lewy FH. Paralysis agitans. In: Pathologische Anatomie.Handbuch der Neurologie, edited by Lewandowsky M.Berlin: Springer-Verlag, 1912, pp. 920–923.

173. Lewy FH. Zur pathologischen anatomie der paralysisagitans. Dtsch Z Nervenheilkd 50: 50–55, 1913.

174. Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ,Lashley T, Quinn NP, Rehncrona S, Bjorklund A, WidnerH, Revesz T, Lindvall O, and Brundin P. Lewy bodies ingrafted neurons in subjects with Parkinson’s disease sug-gest host-to-graft disease propagation. Nat Med 14: 501–503, 2008.

175. Li JY, Henning Jensen P, and Dahlstrom A. Differentiallocalization of alpha-, beta- and gamma-synucleins in therat CNS. Neuroscience 113: 463–478, 2002.

176. Lin MT and Yang JJ. Stimulation of the nigrostriatal do-pamine system produces hypertension and tachycardia inrats. Am J Physiol 266: H2489–H2496, 1994.

177. Linazasoro G. Classical Parkinson disease versus Parkin-son complex—reflections against staging and in favour ofheterogeneity. Eur J Neurol 14: 721–728, 2007.

178. Linthorst AC, van Giersbergen PL, Gras M, Versteeg DH,and de Jong W. The nigrostriatal dopamine system: role inthe development of hypertension in spontaneously hy-pertensive rats. Brain Res 639: 261–268, 1994.

179. Liu G, Zhang C, Yin J, Li X, Cheng F, Li Y, Yang H,Ueda K, Chan P, and Yu S. alpha-Synuclein is differen-tially expressed in mitochondria from different rat brainregions and dose-dependently down-regulates complex Iactivity. Neurosci Lett 454: 187–192, 2009.

180. Loewy AD. Forebrain nuclei involved in autonomic con-trol. Prog Brain Res 87: 253–268, 1991.

181. Lopshire JC and Zipes DP. Device therapy to modulatethe autonomic nervous system to treat heart failure. CurrCardiol Rep 14: 593–600, 2012.

182. Low PA and Bennaroch EE. (Eds). Clinical Autonomic Dis-orders. Philadelphia: Lippincott, Williams, and Wilkins, 2008.

183. Lu SF, Young HJ, and Lin MT. Nigrostriatal dopaminesystem mediates baroreflex sensitivity in rats. NeurosciLett 190: 17–20, 1995.

184. Luk KC, Kehm V, Carroll J, Zhang B, O’Brien P, Tro-janowski JQ, and Lee VM. Pathological alpha-synucleintransmission initiates Parkinson-like neurodegeneration innontransgenic mice. Science 338: 949–953, 2012.

185. Luk KC, Kehm VM, Zhang B, O’Brien P, TrojanowskiJQ, and Lee VM. Intracerebral inoculation of pathologicalalpha-synuclein initiates a rapidly progressive neurode-generative alpha-synucleinopathy in mice. J Exp Med 209:975–986, 2012.

186. Macefield VG and Henderson LA. Real-time imaging ofthe medullary circuitry involved in the generation ofspontaneous muscle sympathetic nerve activity in awakesubjects. Hum Brain Mapp 31: 539–549, 2010.

187. Mahon S, Vautrelle N, Pezard L, Slaght SJ, Deniau JM,Chouvet G, and Charpier S. Distinct patterns of striatalmedium spiny neuron activity during the natural sleep-wake cycle. J Neurosci 26: 12587–12595, 2006.

188. Mallet N, Pogosyan A, Marton LF, Bolam JP, Brown P,and Magill PJ. Parkinsonian beta oscillations in the externalglobus pallidus and their relationship with subthalamicnucleus activity. J Neurosci 28: 14245–14258, 2008.

189. Mallet N, Pogosyan A, Sharott A, Csicsvari J, Bolam JP,Brown P, and Magill PJ. Disrupted dopamine transmissionand the emergence of exaggerated beta oscillations insubthalamic nucleus and cerebral cortex. J Neurosci 28:4795–4806, 2008.

190. Marinella MA. Acute colonic pseudo-obstruction com-plicated by cecal perforation in a patient with Parkinson’sdisease. South Med J 90: 1023–1026, 1997.

191. Martin LJ, Pan Y, Price AC, Sterling W, Copeland NG,Jenkins NA, Price DL, and Lee MK. Parkinson’s diseasealpha-synuclein transgenic mice develop neuronal mito-chondrial degeneration and cell death. J Neurosci 26: 41–50, 2006.

192. Masuda-Suzukake M, Nonaka T, Hosokawa M, OikawaT, Arai T, Akiyama H, Mann DM, and Hasegawa M.Prion-like spreading of pathological alpha-synuclein inbrain. Brain 136: 1128–1138, 2013.

193. McBride PA, Schulz-Schaeffer WJ, Donaldson M, BruceM, Diringer H, Kretzschmar HA, and Beekes M. Earlyspread of scrapie from the gastrointestinal tract to thecentral nervous system involves autonomic fibers of thesplanchnic and vagus nerves. J Virol 75: 9320–9327, 2001.

194. McCoy MK, Martinez TN, Ruhn KA, Szymkowski DE,Smith CG, Botterman BR, Tansey KE, and Tansey MG.Blocking soluble tumor necrosis factor signaling withdominant-negative tumor necrosis factor inhibitor attenu-ates loss of dopaminergic neurons in models of Parkin-son’s disease. J Neurosci 26: 9365–9375, 2006.

DMV IN PD 15

Page 16: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

195. McGeer PL, Schwab C, Parent A, and Doudet D. Presenceof reactive microglia in monkey substantia nigra yearsafter 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine ad-ministration. Ann Neurol 54: 599–604, 2003.

196. Miller N, Noble E, Jones D, and Burn D. Hard to swallow:dysphagia in Parkinson’s disease. Age Ageing 35: 614–618, 2006.

197. Morfini G, Pigino G, Opalach K, Serulle Y, Moreira JE,Sugimori M, Llinas RR, and Brady ST. 1-Methyl-4-phe-nylpyridinium affects fast axonal transport by activationof caspase and protein kinase C. Proc Natl Acad Sci U S A104: 2442–2447, 2007.

198. Morrison LA, Sidman RL, and Fields BN. Direct spreadof reovirus from the intestinal lumen to the central ner-vous system through vagal autonomic nerve fibers. ProcNatl Acad Sci U S A 88: 3852–3856, 1991.

199. Mougenot AL, Bencsik A, Nicot S, Vulin J, Morignat E,Verchere J, Betemps D, Lakhdar L, Legastelois S, andBaron TG. Transmission of prion strains in a transgenicmouse model overexpressing human A53T mutatedalpha-synuclein. J Neuropathol Exp Neurol 70: 377–385,2011.

200. Mougenot AL, Nicot S, Bencsik A, Morignat E, VerchereJ, Lakhdar L, Legastelois S, and Baron T. Prion-like ac-celeration of a synucleinopathy in a transgenic mousemodel. Neurobiol Aging 33: 2225–2228, 2012.

201. Muller T, Erdmann C, Bremen D, Schmidt WE, MuhlackS, Woitalla D, and Goetze O. Impact of gastric emptyingon levodopa pharmacokinetics in Parkinson diseasepatients. Clin Neuropharmacol 29: 61–67, 2006.

202. Neafsey EJ. Prefrontal cortical control of the autonomicnervous system: anatomical and physiological observa-tions. Prog Brain Res 85: 147–165; discussion 165–166,1990.

203. Nicklas WJ, Vyas I, and Heikkila RE. Inhibition ofNADH-linked oxidation in brain mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite of the neurotoxin,1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. Life Sci 36:2503–2508, 1985.

204. Nobrega AC, Rodrigues B, and Melo A. Is silent aspira-tion a risk factor for respiratory infection in Parkinson’sdisease patients? Parkinsonism Relat Disord 14: 646–648,2008.

205. Noorian AR, Rha J, Annerino DM, Bernhard D, TaylorGM, and Greene JG. Alpha-synuclein transgenic micedisplay age-related slowing of gastrointestinal motilityassociated with transgene expression in the vagal system.Neurobiol Dis 48: 9–19, 2012.

206. Nosaka S, Yamamoto T, and Yasunaga K. Localization ofvagal cardioinhibitory preganglionic neurons with ratbrain stem. J Comp Neurol 186: 79–92, 1979.

207. Obeso JA, Jahanshahi M, Alvarez L, Macias R, PedrosoI, Wilkinson L, Pavon N, Day B, Pinto S, Rodriguez-Oroz MC, Tejeiro J, Artieda J, Talelli P, Swayne O,Rodriguez R, Bhatia K, Rodriguez-Diaz M, Lopez G,Guridi J, and Rothwell JC. What can man do withoutbasal ganglia motor output? The effect of combinedunilateral subthalamotomy and pallidotomy in a patientwith Parkinson’s disease. Exp Neurol 220: 283–292,2009.

208. Olofsson PS, Rosas-Ballina M, Levine YA, and TraceyKJ. Rethinking inflammation: neural circuits in theregulation of immunity. Immunol Rev 248: 188–204,2012.

209. Orimo S, Amino T, Itoh Y, Takahashi A, Kojo T, Uchi-hara T, Tsuchiya K, Mori F, Wakabayashi K, and Taka-hashi H. Cardiac sympathetic denervation precedesneuronal loss in the sympathetic ganglia in Lewy bodydisease. Acta Neuropathol 109: 583–588, 2005.

210. Orimo S, Takahashi A, Uchihara T, Mori F, Kakita A,Wakabayashi K, and Takahashi H. Degeneration of car-diac sympathetic nerve begins in the early disease processof Parkinson’s disease. Brain Pathol 17: 24–30, 2007.

211. Orimo S, Uchihara T, Nakamura A, Mori F, Kakita A,Wakabayashi K, and Takahashi H. Axonal alpha-synucleinaggregates herald centripetal degeneration of cardiac sym-pathetic nerve in Parkinson’s disease. Brain 131: 642–650,2008.

212. Parihar MS, Parihar A, Fujita M, Hashimoto M, andGhafourifar P. Mitochondrial association of alpha-synu-clein causes oxidative stress. Cell Mol Life Sci 65: 1272–1284, 2008.

213. Parihar MS, Parihar A, Fujita M, Hashimoto M, andGhafourifar P. Alpha synuclein overexpression and ag-gregation exacerbates impairment of mitochondrial func-tions by augmenting oxidative stress in humanneuroblastoma cells. Int J Biochem Cell Biol 41: 2015–2024, 2009.

214. Park HJ, Lee PH, Ahn YW, Choi YJ, Lee G, Lee DY,Chung ES, and Jin BK. Neuroprotective effect of nicotineon dopaminergic neurons by anti-inflammatory action.Eur J Neurosci 26: 79–89, 2007.

215. Parker WD, Jr., Boyson SJ, and Parks JK. Abnormalitiesof the electron transport chain in idiopathic Parkinson’sdisease. Ann Neurol 26: 719–723, 1989.

216. Parkinson J. An Essay on the Shaking Palsy. London:Whittingham and Rowland, 1817.

217. Parkkinen L, Pirttila T, and Alafuzoff I. Applicability ofcurrent staging/categorization of alpha-synuclein pathol-ogy and their clinical relevance. Acta Neuropathol 115:399–407, 2008.

218. Pathak RU and Davey GP. Complex I and energythresholds in the brain. Biochim Biophys Acta 1777: 777–782, 2008.

219. Patrick A and Epstein O. Review article: gastroparesis.Aliment Pharmacol Ther 27: 724–740, 2008.

220. Pavlov VA, Ochani M, Gallowitsch-Puerta M, Ochani K,Huston JM, Czura CJ, Al-Abed Y, and Tracey KJ. Centralmuscarinic cholinergic regulation of the systemic inflam-matory response during endotoxemia. Proc Natl Acad SciU S A 103: 5219–5223, 2006.

221. Pavlov VA, Parrish WR, Rosas-Ballina M, Ochani M,Puerta M, Ochani K, Chavan S, Al-Abed Y, and TraceyKJ. Brain acetylcholinesterase activity controls systemiccytokine levels through the cholinergic anti-inflammatorypathway. Brain Behav Immun 23: 41–45, 2009.

222. Pavlov VA, Wang H, Czura CJ, Friedman SG, and TraceyKJ. The cholinergic anti-inflammatory pathway: a missinglink in neuroimmunomodulation. Mol Med 9: 125–134,2003.

223. Pazo JH and Belforte JE. Basal ganglia and functions ofthe autonomic nervous system. Cell Mol Neurobiol 22:645–654, 2002.

224. Pazo JH, Gomez-Gonzales M, Tumilasci OR, O’DonnellP, and Murer G. The sialagogue response of striatal do-pamine receptors to L-dopa is not influenced by castrationor chronic estrogen treatment. Brain Res Bull 16: 1–4,1986.

16 GREENE

Page 17: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

225. Pazo JH, Medina JH, and Tumilasci OR. The role of thecaudate-putamen nucleus in salivary secretion induced byL-DOPA. Neuropharmacology 21: 261–265, 1982.

226. Pearce JM. The Lewy body. J Neurol Neurosurg Psy-chiatry 71: 214, 2001.

227. Pfeiffer RF. Gastrointestinal dysfunction in Parkinson’sdisease. Lancet Neurol 2: 107–116, 2003.

228. Pfeiffer RF and Quigley EM. Gastrointestinal motilityproblems in patients with Parkinson’s disease: epidemi-ology, pathophysiology, and guidelines for management.CNS Drugs 11: 435–448, 1999.

229. Phillips RJ, Walter GC, Wilder SL, Baronowsky EA, andPowley TL. Alpha-synuclein-immunopositive myentericneurons and vagal preganglionic terminals: autonomicpathway implicated in Parkinson’s disease? Neuroscience153: 733–750, 2008.

230. Poewe W. Nonmotor symptoms in Parkinson’s disease.In: Parkinson’s Disease and Movement Disorders, editedby Janckovic J and Tolosa E. Philadelphia, PA: Lippin-cott, Wilkins, and Williams, pp. 67–76, 2007.

231. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, De-hejia A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R,Stenroos ES, Chandrasekharappa S, Athanassiadou A,Papapetropoulos T, Johnson WG, Lazzarini AM, DuvoisinRC, Di Iorio G, Golbe LI, and Nussbaum RL. Mutation inthe alpha-synuclein gene identified in families with Par-kinson’s disease. Science 276: 2045–2047, 1997.

232. Pouclet H, Lebouvier T, Coron E, Des Varannes SB,Neunlist M, and Derkinderen P. A comparison betweencolonic submucosa and mucosa to detect Lewy pathologyin Parkinson’s disease. Neurogastroenterol Motil 24:e202–e205, 2012.

233. Powers KM, Kay DM, Factor SA, Zabetian CP, HigginsDS, Samii A, Nutt JG, Griffith A, Leis B, Roberts JW,Martinez ED, Montimurro JS, Checkoway H, and PayamiH. Combined effects of smoking, coffee, and NSAIDs onParkinson’s disease risk. Mov Disord 23: 88–95, 2008.

234. Powley TL and Phillips RJ. Musings on the wanderer:what’s new in our understanding of vago-vagal reflexes? I.Morphology and topography of vagal afferents innervat-ing the GI tract. Am J Physiol Gastrointest Liver Physiol283: G1217–G1225, 2002.

235. Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS,Knapp DJ, and Crews FT. Systemic LPS causes chronicneuroinflammation and progressive neurodegeneration.Glia 55: 453–462, 2007.

236. Qualman SJ, Haupt HM, Yang P, and Hamilton SR.Esophageal Lewy bodies associated with ganglion cellloss in achalasia. Similarity to Parkinson’s disease. Gas-troenterology 87: 848–856, 1984.

237. Quigley EM. Gastric motor and sensory function and motordisorders of the stomach. In: Sleisenger & Fordtran’s Gas-trointestinal and Liver Disease Pathophysiology/Diagnosis/Management, edited by Friedman LS, Brandt LJ, andFeldman M. Philadelphia: Elsevier, 2006, pp. 999–1028.

238. Quik M, Parameswaran N, McCallum SE, Bordia T, BaoS, McCormack A, Kim A, Tyndale RF, Langston JW, andDi Monte DA. Chronic oral nicotine treatment protectsagainst striatal degeneration in MPTP-treated primates. JNeurochem 98: 1866–1875, 2006.

239. Raz A, Feingold A, Zelanskaya V, Vaadia E, and Berg-man H. Neuronal synchronization of tonically activeneurons in the striatum of normal and parkinsonian pri-mates. J Neurophysiol 76: 2083–2088, 1996.

240. Read SJ, Leggett BA, and Pender MP. Gastroparesis withmultiple sclerosis. Lancet 346: 1228, 1995.

241. Reddymasu SC, Bonino J, and McCallum RW. Gastro-paresis secondary to a demyelinating disease: a case se-ries. BMC Gastroenterol 7: 3, 2007.

242. Reynolds AD, Banerjee R, Liu J, Gendelman HE, andMosley RL. Neuroprotective activities of CD4 + CD25 +regulatory T cells in an animal model of Parkinson’sdisease. J Leukoc Biol 82: 1083–1094, 2007.

243. Reynolds AD, Stone DK, Hutter JA, Benner EJ, MosleyRL, and Gendelman HE. Regulatory T cells attenuate th17cell-mediated nigrostriatal dopaminergic neurodegenera-tion in a model of Parkinson’s disease. J Immunol 184:2261–2271, 2010.

244. Rinne OJ, Nurmi E, Ruottinen HM, Bergman J, Eskola O,and Solin O. [(18)F]FDOPA and [(18)F]CFT are bothsensitive PET markers to detect presynaptic dopaminergichypofunction in early Parkinson’s disease. Synapse 40:193–200, 2001.

245. Rizvi SJ, Donovan M, Giacobbe P, Placenza F, RotzingerS, and Kennedy SH. Neurostimulation therapies for treat-ment resistant depression: a focus on vagus nerve stimu-lation and deep brain stimulation. Int Rev Psychiatry 23:424–436, 2011.

246. Rosas-Ballina M, Ochani M, Parrish WR, Ochani K,Harris YT, Huston JM, Chavan S, and Tracey KJ. Splenicnerve is required for cholinergic antiinflammatory path-way control of TNF in endotoxemia. Proc Natl Acad Sci US A 105: 11008–11013, 2008.

247. Rosas-Ballina M, Olofsson PS, Ochani M, Valdes-FerrerSI, Levine YA, Reardon C, Tusche MW, Pavlov VA,Andersson U, Chavan S, Mak TW, and Tracey KJ.Acetylcholine-synthesizing T cells relay neural signals ina vagus nerve circuit. Science 334: 98–101, 2011.

248. Rosas-Ballina M and Tracey KJ. The neurology of theimmune system: neural reflexes regulate immunity. Neu-ron 64: 28–32, 2009.

249. Rosin B, Nevet A, Elias S, Rivlin-Etzion M, Israel Z, andBergman H. Physiology and pathophysiology of the basalganglia-thalamo-cortical networks. Parkinsonism RelatDisord 13 Suppl 3: S437–S439, 2007.

250. Sakakibara R, Shinotoh H, Uchiyama T, Sakuma M,Kashiwado M, Yoshiyama M, and Hattori T. Questionnaire-based assessment of pelvic organ dysfunction in Parkin-son’s disease. Auton Neurosci 92: 76–85, 2001.

251. Sakakibara R, Uchiyama T, Yamanishi T, Shirai K, andHattori T. Bladder and bowel dysfunction in Parkinson’sdisease. J Neural Transm 115: 443–460, 2008.

252. Salat-Foix D, Tran K, Ranawaya R, Meddings J, and Su-chowersky O. Increased intestinal permeability and Parkin-son disease patients: chicken or egg? Can J Neurol Sci 39:185–188, 2012.

253. Samii A, Etminan M, Wiens MO, and Jafari S. NSAID useand the risk of Parkinson’s disease: systematic review andmeta-analysis of observational studies. Drugs Aging 26:769–779, 2009.

254. Schaller BJ, Graf R, and Jacobs AH. Pathophysiologicalchanges of the gastrointestinal tract in ischemic stroke. AmJ Gastroenterol 101: 1655–1665, 2006.

255. Sharott A, Magill PJ, Harnack D, Kupsch A, Meissner W,and Brown P. Dopamine depletion increases the powerand coherence of beta-oscillations in the cerebral cortexand subthalamic nucleus of the awake rat. Eur J Neurosci21: 1413–1422, 2005.

DMV IN PD 17

Page 18: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

256. Siddiqui MF, Rast S, Lynn MJ, Auchus AP, and PfeifferRF. Autonomic dysfunction in Parkinson’s disease: acomprehensive symptom survey. Parkinsonism RelatDisord 8: 277–284, 2002.

257. Singer C, Weiner WJ, and Sanchez-Ramos JR. Autonomicdysfunction in men with Parkinson’s disease. Eur Neurol32: 134–140, 1992.

258. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S,Kachergus J, Hulihan M, Peuralinna T, Dutra A, Nuss-baum R, Lincoln S, Crawley A, Hanson M, MaraganoreD, Adler C, Cookson MR, Muenter M, Baptista M, MillerD, Blancato J, Hardy J, and Gwinn-Hardy K. alpha-Sy-nuclein locus triplication causes Parkinson’s disease.Science 302: 841, 2003.

259. Smith Y, Raju DV, Pare JF, and Sidibe M. The thala-mostriatal system: a highly specific network of the basalganglia circuitry. Trends Neurosci 27: 520–527, 2004.

260. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ,Jakes R, and Goedert M. Alpha-synuclein in Lewy bodies.Nature 388: 839–840, 1997.

261. Sulzer D and Surmeier DJ. Neuronal vulnerability, path-ogenesis, and Parkinson’s disease. Mov Disord 28: 41–50,2013.

262. Tansey MG and Goldberg MS. Neuroinflammation inParkinson’s disease: its role in neuronal death and impli-cations for therapeutic intervention. Neurobiol Dis 37:510–518, 2010.

263. Tansey MG, McCoy MK, and Frank-Cannon TC. Neu-roinflammatory mechanisms in Parkinson’s disease: po-tential environmental triggers, pathways, and targets forearly therapeutic intervention. Exp Neurol 208: 1–25,2007.

264. Taylor TN, Caudle WM, Shepherd KR, Noorian A,Jackson CR, Iuvone PM, Weinshenker D, Greene JG, andMiller GW. Nonmotor symptoms of Parkinson’s diseaserevealed in an animal model with reduced monoaminestorage capacity. J Neurosci 29: 8103–8113, 2009.

265. Teismann P, Tieu K, Choi DK, Wu DC, Naini A, Hunot S,Vila M, Jackson-Lewis V, and Przedborski S. Cycloox-ygenase-2 is instrumental in Parkinson’s disease neuro-degeneration. Proc Natl Acad Sci U S A 100: 5473–5478,2003.

266. Temlett JA and Thompson PD. Reasons for admission tohospital for Parkinson’s disease. Intern Med J 36: 524–526, 2006.

267. Tobi M, Holtz T, Carethers J, and Owyang C. Delayedgastric emptying after laparoscopic anterior highly selec-tive and posterior truncal vagotomy. Am J Gastroenterol90: 810–811, 1995.

268. Travagli RA, Hermann GE, Browning KN, and RogersRC. Brainstem circuits regulating gastric function. AnnuRev Physiol 68: 279–305, 2006.

269. Ueki A and Otsuka M. Life style risks of Parkinson’sdisease: association between decreased water intake andconstipation. J Neurol 251 Suppl 7: vII18–vII23, 2004.

270. Uematsu A, Tsurugizawa T, Uneyama H, and Torii K.Brain-gut communication via vagus nerve modulatesconditioned flavor preference. Eur J Neurosci 31: 1136–1143, 2010.

271. Undem BJ and Carr MJ. Targeting primary afferent nervesfor novel antitussive therapy. Chest 137: 177–184, 2010.

272. Van Laar VS and Berman SB. Mitochondrial dynamics inParkinson’s disease. Exp Neurol 218: 247–256, 2009.

273. Villaran RF, Espinosa-Oliva AM, Sarmiento M, De Pa-blos RM, Arguelles S, Delgado-Cortes MJ, Sobrino V,Van Rooijen N, Venero JL, Herrera AJ, Cano J, andMachado A. Ulcerative colitis exacerbates lipopolysac-charide-induced damage to the nigral dopaminergic sys-tem: potential risk factor in Parkinson‘s disease. JNeurochem 114: 1687–1700, 2010.

274. Vorobyov VV, Schibaev NV, Morelli M, and CartaAR. EEG modifications in the cortex and striatum afterdopaminergic priming in the 6-hydroxydopamine ratmodel of Parkinson’s disease. Brain Res 972: 177–185,2003.

275. Wakabayashi K, Takahashi H, Ohama E, and Ikuta F.Parkinson’s disease: an immunohistochemical study ofLewy body-containing neurons in the enteric nervoussystem. Acta Neuropathol (Berl) 79: 581–583, 1990.

276. Wakabayashi K, Takahashi H, Takeda S, Ohama E, andIkuta F. Parkinson’s disease: the presence of Lewy bodiesin Auerbach’s and Meissner’s plexuses. Acta Neuropathol(Berl) 76: 217–221, 1988.

277. Wakabayashi K, Takahashi H, Takeda S, Ohama E, andIkuta F. Lewy bodies in the enteric nervous system inParkinson’s disease. Arch Histol Cytol 52 Suppl: 191–194,1989.

278. Wakabayashi K, Toyoshima Y, Awamori K, Anezaki T,Yoshimoto M, Tsuji S, and Takahashi H. Restricted oc-currence of Lewy bodies in the dorsal vagal nucleus in apatient with late-onset parkinsonism. J Neurol Sci 165:188–191, 1999.

279. Wang H, Yu M, Ochani M, Amella CA, Tanovic M,Susarla S, Li JH, Wang H, Yang H, Ulloa L, Al-Abed Y,Czura CJ, and Tracey KJ. Nicotinic acetylcholine receptoralpha7 subunit is an essential regulator of inflammation.Nature 421: 384–388, 2003.

280. Weinberger M, Mahant N, Hutchison WD, Lozano AM,Moro E, Hodaie M, Lang AE, and Dostrovsky JO. Betaoscillatory activity in the subthalamic nucleus and its re-lation to dopaminergic response in Parkinson’s disease. JNeurophysiol 96: 3248–3256, 2006.

281. Weller C, Charlett A, Oxlade NL, Dobbs SM, Dobbs RJ,Peterson DW, and Bjarnason IT. Role of chronic infectionand inflammation in the gastrointestinal tract in the eti-ology and pathogenesis of idiopathic parkinsonism. Part 3:predicted probability and gradients of severity of idio-pathic parkinsonism based on H. pylori antibody profile.Helicobacter 10: 288–297, 2005.

282. Whited KL, Hornof WJ, Garcia T, Bohan DC, Larson RF,and Raybould HE. A non-invasive method for measure-ment of gastric emptying in mice: effects of altering fatcontent and CCK A receptor blockade. Neurogas-troenterol Motil 16: 421–427, 2004.

283. Whited KL, Thao D, Lloyd KC, Kopin AS, and RaybouldHE. Targeted disruption of the murine CCK1 receptorgene reduces intestinal lipid-induced feedback inhibitionof gastric function. Am J Physiol Gastrointest LiverPhysiol 291: G156–G162, 2006.

284. Wichmann T, Kliem MA, and Soares J. Slow oscillatorydischarge in the primate basal ganglia. J Neurophysiol 87:1145–1148, 2002.

285. Wichmann T and Soares J. Neuronal firing before andafter burst discharges in the monkey Basal Ganglia ispredictably patterned in the normal state and altered inparkinsonism. J Neurophysiol 95: 2120–2133, 2006.

18 GREENE

Page 19: Causes and Consequences of Degeneration of the Dorsal ... · in the basal ganglia (40, 101, 121, 170, 280). In the parkin-sonian brain, dopaminergic drugs (127, 274) and deep brain

286. Wishart TM, Parson SH, and Gillingwater TH. Synapticvulnerability in neurodegenerative disease. J NeuropatholExp Neurol 65: 733–739, 2006.

287. Woodford H and Walker R. Emergency hospital admis-sions in idiopathic Parkinson’s disease. Mov Disord 20:1104–1108, 2005.

288. Yang H, Antoine DJ, Andersson U, and Tracey KJ. Themany faces of HMGB1: molecular structure-functionalactivity in inflammation, apoptosis, and chemotaxis. JLeukoc Biol 93: 865–873, 2013.

289. Yang H, Lundback P, Ottosson L, Erlandsson-Harris H,Venereau E, Bianchi ME, Al-Abed Y, Andersson U,Tracey KJ, and Antoine DJ. Redox modification of cys-teine residues regulates the cytokine activity of high mo-bility group box-1 (HMGB1). Mol Med 18: 250–259,2012.

290. Yao Z and Wood NW. Cell death pathways in Parkinson’sdisease: role of mitochondria. Antioxid Redox Signal 11:2135–2149, 2009.

291. Zheng LF, Zhang Y, Chen CL, Song J, Fan RF, Cai QQ,Wang ZY, and Zhu JX. Alterations in TH- and ChAT-immunoreactive neurons in the DMV and gastric dys-motility in an LPS-induced PD rat model. Auton Neurosci177: 194–198, 2013.

292. Zibetti M, Torre E, Cinquepalmi A, Rosso M, Ducati A,Bergamasco B, Lanotte M, and Lopiano L. Motor andnonmotor symptom follow-up in parkinsonian patientsafter deep brain stimulation of the subthalamic nucleus.Eur Neurol 58: 218–223, 2007.

Address correspondence to:Dr. James G. Greene

Department of NeurologyEmory University

6009 Woodruff Memorial Research Building101 Woodruff Circle

Atlanta, GA 30322

E-mail: [email protected]

Date of first submission to ARS Central, February 9, 2014;date of acceptance, March 4, 2014.

Abbreviations Used

AS¼ a-synucleinCNS¼ central nervous system

DMV¼ dorsal motor nucleus of the vagus nerveENS¼ enteric nervous system

GI¼ gastrointestinalIBD¼ inflammatory bowel disease

MPTP¼ 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridinePD¼ Parkinson’s disease

PNS¼ peripheral nervous systemRNS¼ reactive nitrogen speciesROS¼ reactive oxygen species

DMV IN PD 19