University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3,...

123
University of Veterinary Medicine Hannover Intravitreal injections in mice as a technique to induce unilateral photoreceptor degeneration in comparison to the genetic model of the rd10 mouse Thesis Submitted in partial fulfilment of the requirements for the degree - Doctor of Veterinary Medicine - Doctor medicinae veterinariae (Dr. med. vet.) by Carina Sarah Rösch Aachen Hannover 2013

Transcript of University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3,...

Page 1: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

University of Veterinary Medicine Hannover

Intravitreal injections in mice as a technique to induce unilateral

photoreceptor degeneration in comparison to the genetic model of

the rd10 mouse

Thesis

Submitted in partial fulfilment of the requirements for the degree

- Doctor of Veterinary Medicine -

Doctor medicinae veterinariae

(Dr. med. vet.)

by

Carina Sarah Rösch

Aachen

Hannover 2013

Page 2: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

Academic supervisors: 1. Univ.-Prof. Dr. med. vet. Christiane Pfarrer

Department of Anatomy

University of Veterinary Medicine Hannover

2. Univ.-Prof. Dr. med. Peter Walter

Department of Ophthalmology

University Hospital RWTH Aachen

1. Referee: Univ.-Prof. Dr. med. vet. Christiane Pfarrer

Department of Anatomy, University of Veterinary Medicine Hannover

Univ.-Prof. Dr. med. Peter Walter

Department of Ophthalmology, University Hospital RWTH Aachen

2. Referee: PD Dr. Veronika M. Stein, PhD, Dipl. ECVN

University of Veterinary Medicine Hannover

Day of oral examination: 12.11.2013

This work was supported by the DFG grants WA 1472/6-1 to PW and MU 3036/3-1 to FM.

Page 3: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

DEDICATED TO MY PARENTS

Page 4: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

Parts of this work were submitted for publication in the following scientific journals:

Graefe´s Archive for Clinical and Experimental Ophthalmology, Springer-Verlag GmbH, Heidelberg,

Germany.

Investigative Ophthalmology & Visual Science (IOVS), ARVO JOURNAL, Rockville, Maryland,

USA.

Journal of Ophthalmology, Hindawi Publishing Corporation, 410 Park Avenue, 15th Floor, #287 pmb,

New York, NY 10022, USA.

Parts of the results of this doctoral thesis were presented as scientific posters (P) or as lecture

(L) at the following symposia and conferences:

Annual Meeting 2013 of the Association for Research in Vision and Ophthalmology (ARVO),

May 05-09, 2013 Seattle, Washington (P):

Effect of Intravitreal Injection of Iodoacetic Acid in Mice as a Model of Pharmacological Induced

Monolateral Photoreceptor Degeneration

Sarah Rösch1,2

, Stephan Hesse1, Christine Haselier

1, Babac A. Mazinani

1, Gernot Roessler

1, Christiane

Pfarrer2, Peter Walter

1

1Department of Ophthalmology, RWTH Aachen University, Germany,

2Department of Anatomy,

University of Veterinary Medicine Hannover, Germany

European Retina Meeting 2013, October 03-05, 2013 Alicante, Spain (P):

Characterisation of selective photoreceptor degeneration in mice induced by intravitreal

injections of N-methyl-N-nitrosourea in comparison to the systemic treatment and a genetic

model.

Sarah Rösch1,2

, Frank Müller3, Christiane Pfarrer

2, Peter Walter

1

1Department of Ophthalmology, RWTH Aachen University, Germany,

2Department of Anatomy,

University of Veterinary Medicine Hannover, Germany, 3Institute of Complex Systems, Cellular

Biophysics, ICS-4, Forschungszentrum Jülich GmbH, Germany

Artificial Vision, November 08-09, 2013 Aachen, Germany (L):

Effects of intravitreal injection of iodoacetic acid and N-methyl-N-nitrosourea on photoreceptor

survival in mice

Sarah Rösch1,2

, Sandra Johnen1, Frank Müller

3, Christiane Pfarrer

2, Peter Walter

1

1Department of Ophthalmology, RWTH Aachen University, Germany,

2Department of Anatomy,

University of Veterinary Medicine Hannover, Germany, 3Institute of Complex Systems, Cellular

Biophysics, ICS-4, Forschungszentrum Jülich GmbH, Germany

Page 5: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

TABLE OF CONTENTS

LIST OF ABBREVIATIONS

1 INTRODUCTION .............................................................................................................. 9

2 LITERATURE SURVEY ................................................................................................ 11

2.1 The eye ..................................................................................................................... 11

2.2 Retinitis pigmentosa ................................................................................................. 11

2.3 Implantable visual prostheses................................................................................... 13

2.4 Genetic animal models of retinal degeneration ........................................................ 14

2.4.1 rd10 mouse ....................................................................................................... 14

2.4.2 Genetic models in larger animals than rodents ................................................ 15

2.5 Induction of photoreceptor degeneration ................................................................. 16

2.5.1 Iodoacetic acid .................................................................................................. 17

2.5.2 N-methyl-N-nitrosourea ................................................................................... 18

2.6 The intravitreal injection .......................................................................................... 18

3 AIM OF THIS THESIS .................................................................................................... 19

4 MATERIALS & METHODS ........................................................................................... 20

4.1 Animals .................................................................................................................... 20

4.2 Intravitreal injections in mice ................................................................................... 22

4.3 Electroretinography .................................................................................................. 23

4.4 Spectral domain Optical Coherence Tomography ................................................... 24

4.5 Fluorescein angiography (FA) ................................................................................. 25

4.6 Histology .................................................................................................................. 25

4.7 Urine test .................................................................................................................. 26

4.8 Statistics ................................................................................................................... 26

5 RESULTS ......................................................................................................................... 27

5.1 STUDY I ......................................................................................................................

Correlations between ERG, OCT and anatomical findings in the rd10 mouse ....... 27

5.1.1 ABSTRACT ..................................................................................................... 28

5.1.2 INTRODUCTION ............................................................................................ 29

5.1.3 MATERIALS AND METHODS ..................................................................... 30

5.1.4 RESULTS ......................................................................................................... 34

5.1.5 DISCUSSION .................................................................................................. 36

5.1.6 CONCLUSIONS .............................................................................................. 37

5.1.7 REFERENCES ................................................................................................. 38

5.1.8 FIGURES ......................................................................................................... 40

Page 6: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

5.2 STUDY II ....................................................................................................................

The effects of iodoacetic acid on the mouse retina .................................................. 45

5.2.1 ABSTRACT ..................................................................................................... 46

5.2.2 INTRODUCTION ............................................................................................ 47

5.2.3 MATERIALS & METHODS ........................................................................... 49

5.2.4 RESULTS ......................................................................................................... 54

5.2.5 DISCUSSION .................................................................................................. 57

5.2.6 ACKNOWLEDGEMENT ............................................................................... 59

5.2.7 REFERENCES ................................................................................................. 60

5.2.8 FIGURES ......................................................................................................... 64

5.3 STUDY III ...................................................................................................................

Selective photoreceptor degeneration by intravitreal injection of MNU ................. 70

5.3.1 ABSTRACT ..................................................................................................... 71

5.3.2 INTRODUCTION ............................................................................................ 72

5.3.3 MATERIALS AND METHODS ..................................................................... 72

5.3.4 RESULTS ......................................................................................................... 80

5.3.5 DISCUSSION .................................................................................................. 84

5.3.6 ACKNOWLEDGEMENT ............................................................................... 87

5.3.7 REFERENCES ................................................................................................. 88

5.3.8 FIGURES ......................................................................................................... 91

6 GENERAL DISCUSSION ............................................................................................. 104

7 SUMMARY ................................................................................................................... 110

8 ZUSAMMENFASSUNG ............................................................................................... 112

9 REFERENCES ............................................................................................................... 114

10 ACKNOWLEDGEMENTS ........................................................................................... 123

Page 7: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

LIST OF ABBREVIATIONS

IVI intravitreal injection

rd10 retinal degeneration; genetic blind mouse strain

RP Retinitis pigmentosa

IAA iodoacetic acid

MNU N-methyl-N-nitrosourea

ERG electroretinogram

sd-OCT spectral domain Optical Coherence Tomography

FA fluorescein angiography

RPE retinal pigment epithelial cells

PRA Progressive retinal atrophy

XLPRA X-linked progressive retinal atrophy

cGMP cyclic guanosine monophosphate

ß-PDE ß-subunit of the rod cGMP phosphodiesterase

P Postnatal days; days after birth

i.v. intravenous

i.p. intraperitoneal

BW body weight

Fig. figure

BAC Bacterial Artificial Chromosome

TG-rabbit transgenic rabbit

GAPDH glyceraldehyd-3-phosphate dehydrogenase

TUNEL terminal deoxynucleotidyl transferase dUTP nick-end labeling staining

AMD age-related macular degeneration

PBS phosphate buffered saline

NaOH sodium hydroxide

DMSO dimethylsulfoxide

PA paraformaldehyde

PB phosphate buffer

GFAP glial fibrillary acidic protein

Page 8: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,
Page 9: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

9

INTRODUCTION

1 INTRODUCTION

In certain diseases of the neural system, the loss of function can be treated by means of

electrical stimulation provided by electronic implants. Concerning the visual system, inherited

retinal degenerations may lead to blindness and are currently not treatable. However, visual

function of impaired patients may be restored by retinal implants (WALTER 2005). Retinitis

pigmentosa (RP) is one example of an inherited human disease with selective photoreceptor

degeneration, in which distinct gene mutations lead to the degeneration of first rods and then

consecutively cones with a peripheral to central gradient, whereas the thickness of the inner

retinal layers remains unaffected (TSUBURA et al. 2010). Animal models for comparable

retinal degenerations are important tools to explore experimental therapies for currently

untreatable conditions. In rodents, genetic models are available resembling photoreceptor

degenerations, such as observed in RP. In mice, for example the retinal degeneration (rd) 10

mutant, carrying a mutation in the gene encoding the ß-subunit of rod cGMP

phosphodiesterase, was described and widely used (CHANG et al. 2007). In these mice,

comparable to RP, a progressive loss of photoreceptors - first rods, then cones - was observed

with increasing age, concomitant with a pronounced reduction in the thickness of the outer

nuclear layer (ONL), whereas the thickness of the inner retinal layers remained unaffected

(CHANG et al. 2007).

However, genetic models of animal species with larger eyes are desirable in order to evaluate

experimental surgical approaches for the treatment of retinal degenerations, but they are

intrinsically expensive and show a very slow disease progression (SCOTT et al. 2011). As

photoreceptor degeneration can also be induced pharmacologically, the aim of this project

was to establish a pharmacological model that avoids systemic side effects together with the

advantage of having an intraindividual control eye. To this end, the - in this context “new” -

technique of intravitreal injections was introduced. The local administration of this injection

technique was hypothesized to induce photoreceptor degeneration similar to the effects

obtained with systemic application, but without affecting the contralateral eye as well as the

general health status of the animal.

Page 10: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

10

INTRODUCTION

In this work, all experiments were performed in mice. The mouse model was used in these

experiments in order to compare the results to the available genetic rd10 mouse model of

retinal degeneration.

After characterisation of wild type as well as rd10 mice using different clinical tools as for

example full-field electroretinogram (ERG), Optical Coherence Tomography (OCT) and

fluorescein angiography in vivo and final histological analysis in vitro, wild type mice were

injected systemically and intravitreally with iodoacetic acid (IAA) as well as N-methyl-N-

nitrosourea (MNU). The effects of these two chemical substances were evaluated by the same

methods (see Figure labelled “Project overview”).

Page 11: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

11

LITERATURE SURVEY

2 LITERATURE SURVEY

2.1 The eye

The eyeball (bulbus oculi) consists of three layers:

Tunica fibrosa bulbi (including sclera and cornea)

Tunica vasculosa bulbi (including choroidea, corpora ciliares and iris)

Tunica nervosa bulbi (retina) (NICKEL et al. 2004)

This hollow sphere, the bulbus oculi, is completed by the lens, the humor aquosus and the

corpus vitreum. Together with the optic nerve, the central visual pathways and the area optica

(visual cortex), they form the sight organ (organum visum) (NICKEL et al. 2004). The tunica

nervosa bulbi consists of the stratum nervosum - with the photoreceptors (rods and cones,

stratum neuroepitheliale), the I. neuron (stratum ganglionare retinae) and the II. neuron

(stratum ganglionare n. optici) - and the stratum pigmentosum retinae (NICKEL et al. 2004).

In humans, the stratum nervosum counts up to 150 million rods and about 6 million cones

(NICKEL et al. 2004). Retinitis pigmentosa (RP) is an inherited retinal disease leading to the

degeneration of photoreceptors and consecutively leading to blindness in humans (TSUBURA

et al. 2010).

The eye of the mouse, the animal species used in these experiments, is organised in the same

way as in humans. The retina is rod dominated with only a few per cent of cones (WANG et

al. 2011). However, the cones are not organized into a central fovea, as in the human eye

(WANG et al. 2011).

2.2 Retinitis pigmentosa

Retinitis pigmentosa (RP) is an inherited, bilaterally progressive, human retinal disease -

while the name “Retinitis pigmentosa”, given by Frans Donders in 1857, indicates

misleadingly an inflammatory disease (TSUBURA et al. 2010). More than 100 different

forms of mutations, mostly missense mutations, which are associated with mutations of the

rod-specific opsin rhodopsin, are leading to the same result of phenotypic Retinitis

pigmentosa (TSO et al. 1997; HARTONG et al. 2006; WANG et al. 2011). RP shows genetic

heterogeneity with autosomal recessive, autosomal dominant and X chromosome-linked traits

(TUNTIVANICH et al. 2009).

Page 12: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

12

LITERATURE SURVEY

During the progress of the disease, first rods and then cones degenerate, because rods seem to

secrete diffusible factors, which are essential for the surviving of the cones (TSO et al. 1997;

HICKS a. SABEL 1999). The interneurons and the ganglion cells of the inner retina remain

relatively intact (SCOTT et al. 2011). A characteristic sign of RP is the intraretinal

pigmentation in bone spicule deposits in the fundus, because the retinal pigment epithelial

cells (RPE) migrate near to the retinal blood vessels, which attenuate (TSUBURA et al.

2010). The prevalence of RP in humans is 1/4000 (TSUBURA et al. 2010). The age of onset

and rate of retinal degeneration varies because of the different forms and the inconsistent

progress; however, symptoms usually begin with loss of night vision in adolescence

(nyctalopia; due to degeneration of first rods), loss of side vision in young adulthood (tunnel

vision; begin of degeneration in peripheral visual field) and loss of central vision later in life

(HARTONG et al. 2006, TSUBURA et al. 2010).

A well-known entity in veterinary medicine is the progressive retinal atrophy (PRA) in dogs,

the canine equivalent of human RP. Rarely, PRA is also seen in the Abyssinian cat (WALDE

et al. 2008). PRA is leading to significant visual impairment, also with different age of onset

(about two years of age) (WALDE et al. 2008). In contrast to RP, in nearly all breeds PRA is

an autosomal recessive form, except the X chromosome linked trait (XLPRA) in the Siberian

Husky and the autosomal dominant form in the Bullmastiff (WALDE et al. 2008). Signs are

hyperreflexitivity of the tapetum lucidum, because of the atrophy of the outer retinal layers, a

thinning of the retinal vessels in the fundus and a brighter optic disc than normal (WALDE et

al. 2008). Hypo- and hyperpigmentation in the tapetum free areas of the fundus lead to a grey

to brown radial striation in later stages (WALDE et al. 2008). Symptoms are firstly nyctalopia

and consecutively blindness as in RP, but in most cases PRA is diagnosed after observation of

a consecutive cataract formation (WALDE et al. 2008).

In humans, no forms of treatment or effective therapies are available for progressive retinal

dystrophies (WALTER 2005). The substitution of vitamin A palmitate and omega-3-rich fish,

retards disease progression (HARTONG et al. 2006). Different research approaches exist to

find possible and successful ways of treatment and to restore sight (SCOTT et al. 2011).

Retinal transplants

Gene therapy

Page 13: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

13

LITERATURE SURVEY

Stem cells

Implantable visual prostheses

2.3 Implantable visual prostheses

Retinal implants are electrical devices in form of microelectrode arrays, which can be fixed

epiretinally (onto the inner layers of the retina),

subretinally (between the inner layers of the retina and the pigment epithelium, at the

place of the destroyed photoreceptors),

around the optic nerve as optic nerve stimulators

or in the region of the visual cortex as cortical prostheses (WALTER 2005).

Implants of all concepts have been implanted in animal experiments as well as in pilot trials in

humans (WALTER 2005). The research group of the University Hospital RWTH Aachen is

working on the epiretinal concept. The epiretinal implant is fixed on the inner surface of the

retina using micro tacks (WALTER 2005). Basically a camera takes pictures of the

environment (FEUCHT 2005). These pictures are processed into spatial and temporal patterns

of stimulation signals and transferred to the chip by telemetry. The chip stimulates the

remaining nerval structures or cells of the retina with an electrical signal. This electrical signal

reaches the visual cortex via the optic nerve and in this manner the chip restores visual

function (FEUCHT 2005).

As one example of an epiretinal implant the ARGUS implant, which was developed in the US

(NATIONAL INSTITUTE FOR HEALTH RESEARCH 2011), has already been transplanted

in human eyes. After the development of the ARGUS I in the year 2002 (16 electrodes; tested

in six patients), now the new ARGUS II (approval 2013; Second Sight medical products,

Sylmar, California, USA) with 60 electrodes is available and can be implanted. This system is

in use in Europe and in the US for treatment of RP. Because it is the only way of RP

treatment, the charges are paid by the health insurance (NATIONAL INSTITUTE FOR

HEALTH RESEARCH 2011).

Another epiretinal implant, the EPIRET-type, was developed in cooperation with the

Department of Ophthalmology of the University Hospital Aachen (ROESSLER et al. 2009;

MENZEL-SEVERING et al. 2012). The microelectrode array of the EPIRET3, as a wireless

device entirely implanted within the eye, consists of 25 electrodes. It was implanted in a

Page 14: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

14

LITERATURE SURVEY

prospective clinical trial into 6 blind RP- patients (ROESSLER et al. 2009; MENZEL-

SEVERING et al. 2012).

Further implants are under development. However, all kinds of implants have to be tested

before they can be implanted in humans. The experimental animals for these trials have to

show the same conditions of selective photoreceptor degeneration, as seen in human RP.

2.4 Genetic animal models of retinal degeneration

In small animals, especially in rodents, genetic models are easily commercially available.

For our study, we used the rd10 mouse to characterize the effects of the selective

photoreceptor degeneration by clinical tools, in comparison to the wild type mouse.

The animal suffering of mice, to get on both eyes completely blind, is maybe not as wearing

as for a larger animal (e.g., rabbit or cat), considering the animal species-specific ability to

suffer (TIERSCHUTZGESETZ 2013). As nocturnal animals, mice have to orientate

predominantly in the dark (ENGELHARDT a. BREVES 2004). Their most important sensory

organs for intraspecies communication and environmental monitoring are the vibrissae or

tactile hairs (AHL 1986), and their smell sense.

The strain and stress of diurnal (active during the day and sleeping at the night) or crepuscular

(active primarily during twilight) animals after development of a complete blindness seems to

be higher, although these animals have other very well developed sensory organs, too

(SMITH a. TAYLOR 1995). In these animal species, a complete blindness would be a higher

load of welfare, than to get blind on only one eye.

2.4.1 rd10 mouse

Rd is the abbreviation of “retinal degeneration” (CHANG et al. 2007). This mutant strain

occurs naturally (HICKS a. SABEL 1999). A missense point mutation in exon 13 of the ß-

subunit of the rod cGMP phosphodiesterase type 6 (ß-PDE) gene leads to the degeneration of

first rods and then consecutively cones with an, in contrast to RP, central to peripheral

gradient (CHANG et al. 2007). The degradation starts postnatally after 16 days (P 16) and the

peak of photoreceptor degeneration is reached at P 25 after birth (CHANG et al. 2007). Chang

et al. (2007) observed the complete loss of photoreceptors on P 60, while the inner retinal

layers survive and the thickness of ganglion cell layer and inner nuclear layer does not

Page 15: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

15

LITERATURE SURVEY

decrease (LIANG et al. 2008; CHANG et al. 2007). The cells of the inner retinal layers, e.g.,

horizontal cells and rod and cone bipolar cells, morphologically change and undergo

remodelling (PHILLIPS et al. 2010).

Beside the rd10 mouse, there are many comparable genetic retinal degeneration models in

mice, e.g., the rd1 mouse. But because of the later onset and milder retinal degeneration in

contrast to the rd1 mouse (degeneration begins at P 8; with 4 weeks of age no photoreceptors

are left), the rd10 mouse provides a better genetic mouse model for human RP (CHANG et al.

2007).

2.4.2 Genetic models in larger animals than rodents

For surgical approaches, only larger animal species such as the pig, the cat, and the rabbit

come into consideration. In all three species genetic engineering and maintaining a transgenic

colony is intrinsically costly and the disease progression is slow (SCOTT et al. 2011).

While the pig is a diurnal animal, the cat and the rabbit are crepuscular animals (SMITH a.

TAYLOR 1995; ENGELHARDT a. BREVES 2004). In housing as laboratory animals and

also as pet, the cat and the rabbit had to change into diurnal as consequence of the

domestication and lighting conditions (SMITH a. TAYLOR 1995).

In general, the pig is an expensive animal and needs a lot of space (RICHTLINIE 2010/63/EU

DES EUROPÄISCHEN PARLAMENTS UND DES RATES 2010). However, the eye is

anatomically remarkably similar to the human eye (SCOTT et al. 2011). The swine retina

contains a visual streak (area centralis), which is cone-dominant, with rod enrichment in the

periphery, reminding of the human retina (WANG et al. 2011). In contrast to humans, this

region is not rod-free (SCOTT et al. 2011). A genetic RP-model of a pig was developed by

TSO et al. (1997). Although the degeneration shows features of human RP, most of the rods

are completely degenerated by nine months (TSO et al. 1997). The long period of time until

the animals express a complete disease pattern in combination with the size of the animals

leads to a difficult handling. In addition, it would be very cost intensive to pursue such a

transgenic colony (WANG et al. 2011).

The cat is used for retinal implant research in Australia. There are high demands regarding the

way these animals are kept (SMITH a. TAYLOR 1995). An important model of human

Retinitis pigmentosa in cats has been maintained for more than 28 years by Kristina

Page 16: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

16

LITERATURE SURVEY

Narfström (MENOTTI-RAYMOND et al. 2007). But also as seen in the pig, the complete

photoreceptor degeneration and blindness is observed after a longer time period of disease

progression, in this case with the age of three to five years (MENOTTI-RAYMOND et al.

2007).

The husbandry conditions for rabbits are more easily to fulfil (SMITH a. TAYLOR 1995). In

rabbits one genetic strain was generated in Nagoya, Japan, by bacterial artificial chromosome

(BAC) transgenesis in the year 2008. This first genetic model of photoreceptor degeneration -

the transgenic (TG) rabbit - exhibited rod-dominant, progressive photoreceptor degeneration

(KONDO et al. 2009). But also in this genetic animal model, it would be extremely expensive

to get a colony of genetic rabbits to work on.

It would be useful to have the possibility to induce in reproducible manner photoreceptor

degeneration in any wanted animal species.

2.5 Induction of photoreceptor degeneration

Different physical and chemical methods are described to induce photoreceptor degeneration.

In comparison to the genetic models, they are relatively rapid and cost-effective alternatives

and can be performed in any wanted animal species (WANG et al. 2011). But only in a very

few reports describing the induction of outer retinal degenerations, the well-being, or the

effect on the health status of the experimental animals, was under consideration. In most

cases, the experiments only took a few days, and the long term effects were not seen or

evaluated.

Light exposure is a physical method and is mainly successful in albino animals. The melanin

in the pigment epithelium of pigmented eyes seems to protect against this kind of damage

(RAPP a. WILLIAMS 1980). Even if the eyes of pigmented animals are dilated, the

destruction of the photoreceptors takes longer compared to albinos, due to the absorption of

light by the pigment epithelium (RAPP a. WILLIAMS 1980). Continuous light, also with a

low light intensity, can be a toxic agent (RAPP a. WILLIAMS 1980). Animals have to be

kept in husbandry conditions since birth with very low LUX values (brightness value) or dim

cyclic light (RAPP a. WILLIAMS 1980) - so an own breeding not with the commercial high

Page 17: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

17

LITERATURE SURVEY

LUX values is assumed. The prolonged light exposure in animals led to weight loss of 20%,

possibly caused by stress (DUREAU et al. 1996).

Outer retinal degeneration can also be induced chemically. An example is the implantation of

iron particles in the vitreous cavity (WANG et al. 1998). The cause for the iron-induced

retinopathy is an apoptotic mechanism limited to the outer nuclear layer (WANG et al. 1998).

This disease pattern is known as “siderosis bulbi” in human medicine (TERAI et al. 2011).

Beside the effects on the retina, also side effects on the iris (heterochromia, pupilloplegia) and

a shift of the lens were observed (TERAI et al. 2011).

Further the systemic application of special pharmaceuticals leads to a selective photoreceptor

degeneration. These drugs are used since about 50 years in nearly all kinds of animals. Given

systemically they have considerable systemic toxic side effects, e.g., inhibition of glycolysis

(LIANG et al. 2008) and the induction of tumours (SCHREIBER et al. 1969). Some

pharmaceuticals, e.g., sodium iodate, which leads to photoreceptor degeneration by

destruction of the retinal pigment epithelium (WANG et al. 2011), are already lethal in larger

animals such as the pig in concentrations, which have not yet a toxic effect on the retina

(NOEL et al. 2012).

2.5.1 Iodoacetic acid

Iodoacetic acid (IAA) is a photoreceptor toxin, which leads to selective photoreceptor

degeneration when administered systemically (ORZALESI et al. 1970). The inner retinal

neurons remain intact (WANG et al. 2011). The effect is described in many animal species.

IAA reacts with thiol (SH) groups, imidazole and amino groups of proteins, so that toxic side

effects after systemic application are likely. Beside the irreversible inhibition of the thiol (SH)

group of enzymes, as e.g., in the glutathione reductase (MATSUMOTO et al. 2002), IAA

inhibits the glyceraldehyde-3-phosphate dehydrogenase (GAPDH), an enzyme, which is very

important for the metabolic energy production in the glycolysis (LIANG et al. 2008, WANG

et al. 2011). Photoreceptors have a high metabolic rate and therefore seem to be particularly

sensitive for IAA (WANG et al. 2011).

However, after systemic intravenous treatment, not only the retinal enzymes are affected. The

toxic systemic side effects of IAA are reported to lead to a highly reduced animal welfare and

Page 18: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

18

LITERATURE SURVEY

to mortality, e.g., 20% in the experiments with rabbits by ORZALESI et al. (1970) and 50%

in experiments with rats by GRAYMORE et al. (1959).

In other reports or animal species the systemic side effects on the well-being, e.g. weight loss,

were not mentioned.

2.5.2 N-methyl-N-nitrosourea

N-methyl-N-nitrosourea (MNU) is an alkylating agent, belonging to the group of the

nitrosoureas (TSUBURA et al. 2003). Like the other chemical agents of this group, it is toxic,

carcinogenic, teratogenic, embryotoxic and mutagenic (SIGMA-ALDRICH 2013). After

systemic application (i.v.) to rabbits, it leads in 70% to brain tumours and tumours of the

medulla (SCHREIBER et al. 1969).

Herrold (1967) first described that the systemic application in a variety of animals induces

photoreceptor degeneration, while inner neurons remain intact (HERROLD 1967, TSUBURA

et al. 2010). MNU leads to these retinal changes seven days after a single injection by an

apoptotic mechanism as verified by terminal deoxynucleotidyl transferase dUTP nick-end

labelling staining (TUNEL), which is a marker for apoptotic cells (TSURUMA et al. 2012).

The degradation of photoreceptors was due to oxidative stress (TSURUMA et al. 2012).

However, comparable to the systemic administration of IAA, there are also systemic side

effects.

Because of its carcinogenic properties, the user has to follow many safety measures

(protective clothing, etc.), comparable to the use of other cytostatic drugs, as also seen in the

veterinary medicine (HEINEMANN a. MEICHNER 2011).

2.6 The intravitreal injection

The intravitreal injection is a technique to bring drugs for the therapy of eye disease very

close to the area, where they are supposed to act. Under local anaesthesia the drug is placed

with an injection cannula directly into the vitreous humour of the eye (BERUFSVERBAND

DER AUGENÄRZTE DEUTSCHLANDS E.V., DEUTSCHE OPHTHALMOLOGISCHE

GESELLSCHAFT (DOG) 2010). Whereas a decade ago, intravitreal injections were only

used in sporadic, mainly urgent cases, nowadays this technique is very commonly used due to

the introduction of new drugs for the treatment of age-related macular degeneration (AMD)

Page 19: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

19

AIM OF THIS THESIS

and other macular diseases (BERUFSVERBAND DER AUGENÄRZTE DEUTSCHLANDS

E.V., DEUTSCHE OPHTHALMOLOGISCHE GESELLSCHAFT (DOG) 2010).

3 AIM OF THIS THESIS

The aim of this project was to establish a pharmacological model of selective photoreceptor

degeneration in mice by intravitreal injection that avoids systemic side effects combined with

the advantage of having an intraindividual control eye.

Page 20: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

20

MATERIALS & METHODS

4 MATERIALS & METHODS

4.1 Animals

The materials and methods were used and performed as described in detail in the referring

manuscripts (Study I: ”Correlations between ERG, OCT and anatomical findings in the rd10

mouse”; Study II: “The effects of iodoacetic acid on the mouse retina”, Study III: “Selective

photoreceptor degeneration by intravitreal injection of N-methyl-N-nitrosourea”).

All experiments were performed in accordance with the ARVO Statement for the use of

animals in ophthalmic and vision research and in accordance with the German Law for the

Protection of Animals and after approval was obtained by the regulatory authorities. The

proposal of the animal experiments (Study I, II, III) was approved by the LANUV

(Landesamt für Natur, Umwelt und Verbraucherschutz Nordrhein Westfalen, Recklinghausen,

Germany; file reference: 84-02.04.2011.A386).

All pigmented wild type mice (C57BL/6J) and rd10 mice were housed in typical cages with

food and water accessible ad libitum. The room was kept under a 12 h: 12 h light-dark cycle.

C57BL/6J wild type mice and rd10 mice were examined and compared at the age of 2, 3, 5, 7,

9, 12, 24, and 48 weeks (each age group n=3) using full-field electroretinography (ERG),

spectral domain Optical Coherence Tomography (sd-OCT), fluorescein angiography (FA),

Hematoxylin & Eosin histology (HE) and immunohistochemistry (IH).

Wild type mice (C57BL/6J) were treated with iodoacetic acid (IAA) or N-methyl-N-

nitrosourea (MNU). The animals were examined using full-field electroretinogram (ERG) and

spectral domain Optical Coherence Tomography (sd-OCT) five days before, as well as 7 days

and 14 days after injection of IAA and MNU, to monitor photoreceptor function and effects of

the injection. For the duration of experiments, body weight was daily recorded. Finally, two

weeks after treatment eyes were examined in histology (HE and IH).

Page 21: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

21

MATERIALS & METHODS

A total of 20 wild type mice (C57BL/6J; 6 to 8 weeks) received systemically

(intraperitoneally and intravenously) sterile IAA (Sigma Aldrich, Germany). IAA was

dissolved just before use in sterile phosphate buffered saline (PBS) and the pH was adjusted

with NaOH to the physiological pH of 7.4.

Eight animals were treated with an intraperitoneal injection of IAA: 40 mg/kg BW (n=1), 50

mg/kg BW (n=1), 60 mg/kg BW (n=2), 65 mg/kg BW (n=1), 70 mg/kg BW (n=1), 75 mg/kg

BW (n=1) and 2 x 30 mg/kg BW in a time interval of 3.5 hours (n=1).

Twelve animals received intravenous injections of IAA: 55 mg/kg BW (n=2), 60 mg/kg BW

(n=5), 65 mg/kg BW (n=1) and 2 x 30 mg/kg BW in a time interval of 3.5 hours (n=4).

(Study II: “The effects of iodoacetic acid on the mouse retina”)

Three wild type mice (C57BL/6J; 6 to 8 weeks) received intraperitoneally sterile MNU

(Sigma Aldrich, Germany; 60 mg/kg BW). The stability of MNU is reported to be poor in

aqueous solutions and the duration of the procedure of injection is comparably long.

Therefore, MNU was dissolved in dimethylsulfoxide (DMSO; Serva, Heidelberg, Germany)

immediately before use and further diluted (sterile phosphate buffered saline, PBS). Injections

were slowly performed. Temperature of the solution was room temperature.

Animals were kept in one-way cages under a fume hood after the application of MNU (PET;

Tecniplast Deutschland GmbH; Hohenpreißenberg; Germany) under the same housing

conditions as before, because of the possible environmental hazard due to the carcinogenic

and teratogenic potential of N-methyl-N-nitrosourea. To evaluate possible systemic effects of

changing the cage conditions from normal cages to the one-way system, two untreated adult

pigmented wild type mice were kept under the same husbandry conditions (Study III:

“Selective photoreceptor degeneration by intravitreal injection of N-methyl-N-nitrosourea”).

Control animals received systemic sham injections (PBS intraperitoneal (n=3) and

intravenous (n=3) or DMSO + PBS intraperitoneal (n=3)). (Study II: “The effects of

iodoacetic acid on the mouse retina”, Study III: “Selective photoreceptor degeneration by

intravitreal injection of N-methyl-N-nitrosourea”)

Page 22: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

22

MATERIALS & METHODS

For all experimental procedures and examinations (excluding the intraperitoneal application

of MNU and IAA, and the intravenous application of IAA), the animals were anaesthetised

with an intraperitoneal injection of ketamine (70 mg/kg Ketamin® 10%, CEVA, Germany)

and xylazine (10 mg/kg Xylazin 2% Bernburg®, Medistar, Ascheberg, Germany) in 0.1 mL

of saline. (Study I: ”Correlations between ERG, OCT and anatomical findings in the rd10

mouse”; Study II: “The effects of iodoacetic acid on the mouse retina”, Study III: “Selective

photoreceptor degeneration by intravitreal injection of N-methyl-N-nitrosourea”)

4.2 Intravitreal injections in mice

After anaesthesia by intraperitoneal injection of a mixture of ketamine (70 mg/kg Ketamin®

10%, CEVA, Germany) and xylazine (10 mg/kg Xylazin 2% Bernburg®, Medistar,

Ascheberg, Germany) in 0.1 mL of saline, the left eye was locally anaesthetised with

proxymetacainhydrochloride 0.5% eye drops (Proparakain-POS®, Ursapharm, Saarbrücken,

Germany).

A nanoliter injection system and a 3D micromanipulator (Nanoliter 2000; World Precision

Instruments, Inc. Sarasota, FL, USA) were used to conduct intravitreal injections of a

standard volume of 2 µL. After puncture in the dorso-nasal area of the limbus by a 27 gauge

steel needle (BD Microlance TM

3, Heidelberg, Germany) the volume was injected with a

glass capillary in an angle of 30°. Preliminary experiments with control animals (n=9) showed

that the injection technique worked and a volume of 2 µL could be placed safely in the

vitreous cavity of mice. As control intervention 2 µL PBS (n=3) and 2 µL PBS + DMSO

(n=3) were injected each into three eyes intravitreally. Intravitreal injections of fluorescein

(different concentrations dissolved in PBS, n=3) were performed to confirm the safety of the

injection technique. Therefore, images were taken directly after injection of fluorescein by a

confocal laser scanning microscope (Heidelberg Retina Angiograph-1, Heidelberg

Engineering, Germany). (Study II: “The effects of iodoacetic acid on the mouse retina”, Study

III: “Selective photoreceptor degeneration by intravitreal injection of N-methyl-N-

nitrosourea”)

A standard volume of 2µL with different IAA (dissolved in PBS) and MNU (dissolved in

DMSO + PBS) concentrations was injected intravitreally in the left eye of a total of 49 mice.

Page 23: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

23

MATERIALS & METHODS

25 mice were injected with IAA: 0.1 mg/kg BW (n=3), 0.15 mg/kg BW (n=5), 0.2 mg/kg BW

(n=6), 0.25 mg/kg BW (n=4), 0.5 mg/kg BW (n=4), 1 mg/kg BW (n=3). (Study II: “The

effects of iodoacetic acid on the mouse retina”)

24 mice were injected intravitreally with MNU: 0.15 mg/kg BW (n=3), 0.2 mg/kg BW (n=3),

0.25 mg/kg BW (n=3), 0.3 mg/kg BW (n=3), 0.5 mg/kg BW (n=3), 1.0 mg/kg BW (n=3), 2.0

mg/kg BW (n=3), 3.0 mg/kg BW (n=3). (Study III: “Selective photoreceptor degeneration by

intravitreal injection of N-methyl-N-nitrosourea”)

After intravitreal injections antibiotic eye ointment (Polyspectran, Alcon Pharma GmbH,

Freiburg, Germany) was used for three days after application. All intravitreally injected

animals were examined using the same methods (ERG, OCT, Histology) and time schedule as

systemically injected animals. (Study II: “The effects of iodoacetic acid on the mouse retina”,

Study III: “Selective photoreceptor degeneration by intravitreal injection of N-methyl-N-

nitrosourea”)

4.3 Electroretinography

The ERG reveals the electrical activity of the different cell types of the retina, as the answer

to a light stimulus. The ERG wave, as a summarized potential, consists of a first negative

deflection or a-wave (mainly build by rod and cone activity) and a following positive b-wave

(formed by Müller cells and bipolar cells) (KRISHNA et al. 2002). As a non-invasive clinical

tool, it is very useful to monitor the severity and progression of retinal degeneration

(KRISHNA et al. 2002).

Before the electroretinogram procedure, mice were dark adapted (1 hour) and anaesthetised

under red dim light. Pupils were dilated using Tropicamide 2.5% eye drops (Pharmacy of the

University Hospital Aachen, Germany). Proxymetacainhydrochloride 0.5% eye drops

(Proparakain-POS®, Ursapharm, Saarbrücken, Germany) were used for local anaesthesia, and

methylcellulose (Methocel® 2%, Omni Vision, Puchheim, Germany) served for a better

contact and protected the eye against drying out. As active electrodes goldring electrodes

(animal electrode 0.5mm ø 3mm Roland Consult, Brandenburg, Germany) were placed on the

Page 24: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

24

MATERIALS & METHODS

left and right eye. As reference, a goldring electrode was placed in direct contact to the mouth

mucosa. A subcutaneous needle electrode in the lumbar region served as ground electrode.

Ganzfeld (full-field) electroretinogram recordings (ERG) were performed according to the

ISCEV standard protocol (MARMOR et al. 2008) with the Reti System designed for rodents

(Roland Consult Electrophysiological Diagnostic Systems, Brandenburg, Germany). A- and

b-wave amplitudes and implicit times of rod and cone responses were analyzed after

averaging five responses to light stimulation. (Study I: ”Correlations between ERG, OCT and

anatomical findings in the rd10 mouse”; Study II: “The effects of iodoacetic acid on the

mouse retina”, Study III: “Selective photoreceptor degeneration by intravitreal injection of

N-methyl-N-nitrosourea”)

4.4 Spectral domain Optical Coherence Tomography

Optical coherence tomography (OCT) is a non-invasive high-resolution technique to perform

cross-sectional images of the retina in vivo. The technique is comparable to ultrasound, but

instead of using sound the OCT is an interferometric technique using light (YAMAUCHI et

al. 2012).

Spectral domain Optical Coherence Tomography (sd-OCT) scans were performed with the

Spectralis® OCT system (Heidelberg Engineering, Heidelberg, Germany) and a special lens

(+25 D lens) in front of the scanning system, to correct for rodent optics in mice. Artificial

tears (methylcellulose, Methocel® 2%, Omni Vision, Puchheim, Germany) were used

throughout the whole procedure to maintain corneal clarity. The retina was evaluated by cross

sectional images centered on the optic disk, as the main landmark and the confocal image of

the fundus was recorded and examined (IR reflection image, wavelength 715nm). Retinal

thickness was measured at six positions across the retina. For each eye these thickness values

were averaged. The thickness of the different layers of the retina was determined in the same

way. A Nikon D80 camera was used for macroscopic pictures (Nikon AF Micro Nikkor 105

mm, 2896 Pixel x 1944 Pixel x 24 Bit). (Study I: ”Correlations between ERG, OCT and

anatomical findings in the rd10 mouse”; Study II: “The effects of iodoacetic acid on the

mouse retina”, Study III: “Selective photoreceptor degeneration by intravitreal injection of

N-methyl-N-nitrosourea”)

Page 25: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

25

MATERIALS & METHODS

4.5 Fluorescein angiography (FA)

Retinal vasculature was evaluated in anaesthesia and after the dilatation of the pupils by

Tropicamide 2.5% eye drops (Pharmacy of the University Hospital Aachen, Germany) by the

intraperitoneal application of fluorescein (10-20 µL, Fluorescein Alcon® 10%, Alcon Pharma

GMBH, Freiburg im Breisgau, Germany). Images were taken by a confocal laser scanning

microscope (Heidelberg Retina Angiograph-1, Heidelberg Engineering, Germany) in the

fluorescein angiography mode. (Study I: ”Correlations between ERG, OCT and anatomical

findings in the rd10 mouse”)

4.6 Histology

Finally, mice were euthanized by isoflurane overdosing (Forene 100% (V/V)®, Abott GmBH,

Wiesbaden, Germany), decapitated and enucleated. Histology was performed as described

earlier by Mataruga et al. (2007) and in the following three studies.

In brief, after fixation, slices were incubated in blocking solution followed by overnight

incubation in primary antibodies diluted in the same blocking solution. Primary antibodies

used were raised against glutamine synthetase (raised in mouse, 1:4000, BD Biosciences,

Germany); against GFAP (anti glial fibrillary acidic protein, raised in chicken, 1:2000, Novus,

Germany); against CabP (anti calbindin 28K, raised in mouse, 1:1000, Sigma; Germany);

against HCN1 (RTQ-7C3, raised in rat, 1:10, F. Müller, Forschungszentrum Jülich,

Germany); against PKC (anti protein kinase C α, raised in rabbit, 1:4000, Santa Cruz, USA);

against rhodopsin (1D4, 1:500, R.S. Molday, British Columbia, Canada); against calretinin

(AB1550, raised in goat, 1:3000, Millipore, Germany); against recoverin (AB5585, raised in

rabbit, 1:2000, Millipore, Germany). Sections were washed in PB and incubated with

secondary antibodies. Secondary antibodies included: donkey anti-mouse Cy3 (1:100,

Dianova, Germany); donkey anti-rat Cy3 (1:500, Dianova, Germany); donkey anti-chicken

Cy2 (1:400, Dianova, Germany); donkey anti-rabbit Cy2 (1:400, Dianova, Germany); donkey

anti-goat Alexa647 (1:200, Invitrogen, Germany); donkey anti-mouse Dy649 (1:500,

Dianova, Germany) and finally the lectin Peanut agglutinin (PNA, biotinylated, 1:1600,

Sigma Aldrich, Germany) was visualized using StreptavidinAlexa647 (1:100, Invitrogen,

Germany). Imaging was performed with a Leica TCS confocal laser scanning microscope

(Leica Microsystems, Heidelberg, Germany).

Page 26: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

26

MATERIALS & METHODS

For Hematoxylin-Eosin staining, eyes were punctured three times at the limbus and fixated.

After dehydration and incubation in a series of increasing ethanol concentrations, followed by

xylene and paraffin, eyes were embedded in paraffin. Eyes were cut with a microtome (R.

Jung, Heidelberg, Germany), collected on slides and deparaffinised. After rehydration, slices

were stained with Hematoxylin and Eosin and images were performed with a Leica DMRX

microscope (Leica Microsystems, Heidelberg, Germany).

For thickness evaluation comparable to the OCT the papilla was used as landmark. Thickness

was determined in six positions close to the papilla and values were averaged.

(Study I: ”Correlations between ERG, OCT and anatomical findings in the rd10 mouse”;

Study II: “The effects of iodoacetic acid on the mouse retina”, Study III: “Selective

photoreceptor degeneration by intravitreal injection of N-methyl-N-nitrosourea”)

4.7 Urine test

In the first experiments with MNU injections in mice, before, two and three hours, one and

two days after systemic and intravitreal application of MNU urinalyses by UPLC were

performed (Ultra performance liquid chromatography (UPLC), Aquity, Waters). The

excretion of MNU was tested for safety reasons. (Study III: “Selective photoreceptor

degeneration by intravitreal injection of N-methyl-N-nitrosourea”)

4.8 Statistics

Retinal thickness and the thickness of different retinal layers were determined in cross

sectional images of the OCT and in histology by six vertical cuts through the retina in the

height of the papilla. The values were averaged. In the ERG a- and b-wave amplitudes and

implicit times of rod and cone responses were determined.

Statistical analysis of all experiments was performed using one-way ANOVA with

Bonferroni’s post hoc test (GraphPad Prism 5; GraphPad Software, La Jolla, CA). Values

represent mean + standard deviation.

Page 27: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

27

RESULTS

5 RESULTS

5.1 STUDY I

Correlations between ERG, OCT and anatomical

findings in the rd10 mouse

Sarah Rösch1,2

, Sandra Johnen1, Frank Müller

3, Christiane Pfarrer

2, Peter Walter

1

SUBMITTED/ UNDER REVIEW

1. Department of Ophthalmology, RWTH Aachen University, Germany

2. Department of Anatomy, University of Veterinary Medicine Hannover, Germany

3. Institute of Complex Systems, Cellular Biophysics, ICS-4, Forschungszentrum Jülich

GmbH, Germany

Corresponding Author: Peter Walter, Department of Ophthalmology, RWTH Aachen

University, Pauwelsstr. 30, 52074 Aachen, Tel +49-241-8088191, fax +49-241-8082408,

mailto [email protected]

This work is part of a doctoral thesis at the University of Veterinary medicine Hannover

(October 2013).

Keywords: retinal degeneration, rd10 mouse

Word count: 2884

This work was supported by the DFG grants WA 1472/6-1 to PW and MU 3036/3-1 to FM.

Page 28: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

28

RESULTS

5.1.1 ABSTRACT

Background To evaluate the correlation between ERG, OCT, and microscopic findings in the

rd10 mouse.

Methods C57BL/6J wild type mice and rd10 mice were compared at the age of 2, 3, 5, 7, 9,

12, 24, and 48 weeks (each age group n=3) using full-field electroretinography (ERG),

spectral domain Optical Coherence Tomography (sd-OCT), fluorescein angiography (FA),

Hematoxylin & Eosin histology (HE), and immunohistology (IH).

Results While in wild type mice the amplitude of a- and b-wave increased with light intensity

and with the age of the animals, the rd10 mice showed an extinction of the ERG beginning

with the age of 5 weeks. In OCT recordings the thickness of the retina decreased up to 9

weeks of age, mainly based on the degradation of the outer nuclear layer (ONL). Afterwards,

the ONL was no longer visible in the OCT. HE staining and immunohistological findings

confirmed the in vivo data.

Conclusion ERG and OCT are useful methods to evaluate the retinal function and structure in

vivo. The retinal changes seen in the OCT closely match those observed in histological

stainings.

Page 29: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

29

RESULTS

5.1.2 INTRODUCTION

Hereditary dystrophies of the retina, such as Retinitis pigmentosa (RP), are considerable

causes of blindness in humans.[1] Research efforts concerning these currently not treatable

diseases are focused on the genetic background, the mechanisms of degeneration and on

possible treatment strategies.[2] Transgenic animal models in rodents, e.g., the retinal

degeneration (rd) 1 and rd10 mice are well characterized and described.[3-9] In rd1 and rd10

mice, missense point mutations in the gene encoding for the ß-subunit of rod cGMP

phosphodiesterase type 6 (ßPDE) result in the described disease pattern.[3-9] The gene

mutation leads to the degeneration of first rods and then consecutively cones with a central to

peripheral gradient.[9] The fact that in both animal models firstly rods and secondly cones

degenerate, which is comparable to RP,[3-9] seems to be caused by missing diffusible factors

normally secreted by the rods.[10] This kind of retinal degeneration is concomitant with a

pronounced reduction in the thickness of the outer nuclear layer (ONL).[3-9] The thickness of

the inner retinal layers remains nearly unaffected.[3-9] The retinal development in the rd1 and

rd10 mice is comparable to normal mice at 8 postnatal days (P).[2] However, while in the rd1

mice the degeneration becomes apparent with P11, in rd10 mice the degeneration starts at P16

and the peak of photoreceptor degeneration is reached at P25.[9] By P60, no photoreceptors

are left.[9]

Because of the later onset and milder retinal degeneration, the rd10 mice seem to be more

suitable to study slow disease mechanisms in RP.[8] Retinal vessels become sclerotic at 4

weeks of age, the a-wave and b-wave in the electroretinogram are visible with the age of 3

weeks and no longer detectable with the age of 2 months.[8]

To characterize retinal degeneration, monitoring of the time course of the disease is

indispensible. Using histology to observe retinal changes requires a large number of animals

to examine. Therefore it is of interest, if data from functional examinations and in vivo

imaging correlate well with histological data. We examined eyes of wild type and rd10 mice

in different age groups to determine retinal function by means of the full-field

electroretinogram (ERG) and retinal thickness by means of spectral domain Optical

Coherence Tomography (sd-OCT) scanning. The ERG examinations were performed in

general anaesthesia with ketamine and xylazine, because anaesthesia was reported to have no

influence, neither on the oscillatory potentials nor on the ERG waveforms.[11]

Page 30: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

30

RESULTS

5.1.3 MATERIALS AND METHODS

The materials and methods were performed as also described in Study II: “The effects of

iodoacetic acid on the mouse retina” and Study III: “Selective photoreceptor degeneration by

intravitreal injection of N-methyl-N-nitrosourea”.

All experiments were performed in accordance with the ARVO Statement for the use of

animals in ophthalmic and vision research and in accordance with the German Law for the

Protection of Animals and after approval was obtained by the regulatory authorities. All

possible steps were taken to avoid animal suffering at each stage of the experiment. The

proposal of the animal experiments was approved by the LANUV (Landesamt für Natur,

Umwelt und Verbraucherschutz Nordrhein Westfalen, Recklinghausen, Germany; file

reference: 84-02.04.2011.A386).

2.1. Animals

Adult pigmented wild type mice (C57BL/6J) and rd10 mice were maintained under controlled

light conditions (12:12 hours light/dark cycle) with food and water available ad libitum.

Animals (n=3 in each group) were examined at the age of 2, 3, 5, 7, 9, 12, 24, and 48 weeks.

Animals of the same age were sacrificed for histological examinations.

For ERG, OCT, and fluorescein angiography, animals were anaesthetized with an

intraperitoneal injection of a mixture of ketamine (70 mg/kg Ketamin® 10%, CEVA,

Germany) and xylazine (10 mg/kg Xylazin 2% Bernburg®, Medistar, Ascheberg, Germany)

in 0.1 mL of saline.

2.2. Electroretinogram recordings

Electroretinogram recordings (ERG) were performed with the Reti System designed for

rodents (Roland Consult Electrophysiological Diagnostic Systems, Brandenburg, Germany).

Mice were dark adapted for one hour and the pupils were dilated using 2.5% phenylephrine

hydrochloride ophthalmic solution (Tropicamide 2.5% eye drops, Pharmacy of the University

Hospital Aachen, Germany). After local anaesthesia with proxymetacainhydrochloride 0.5%

eye drops (Proparakain- POS®, Ursapharm, Saarbrücken, Germany), a custom-made goldring

Page 31: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

31

RESULTS

electrode (animal electrode 0.5mm ø 3mm Roland Consult) was placed on the corneal surface

of each eye. Methylcellulose (Methocel ® 2%, Omni Vision, Puchheim, Germany) served for

a good contact and to maintain corneal moisture. The reference goldring electrode was placed

on the mouth mucosa and a subcutaneous silver needle electrode in the lumbar region served

as ground electrode. The ERG was recorded as full-field ERGs according to the ISCEV

standard protocol.[12] Five responses to light stimulation were averaged. A- and b-wave

amplitudes and implicit times of rod and cone responses were analyzed.

2.3. Spectral domain Optical Coherence Tomography

Spectral domain Optical Coherence Tomography (sd-OCT) scans were done using the

Spectralis® OCT system (Heidelberg Engineering, Heidelberg, Germany). To correct for

rodent optics, the system was modified according to recommendations of the manufacturer

with a +25 D lens in front of the scanning system. Scans were obtained from the retina

centered on the optic disk as the main landmark. Retinal thickness was measured at six

positions across the retina and averaged for each eye. The thickness of the different retinal

layers was determined using the same technique. The confocal image (IR reflection image,

wavelength 715nm) of the fundus recorded by the OCT system was used for documentation.

2.4. Fluorescein Angiography

Fluorescein angiography was performed to evaluate the retinal vasculature. After dilatation of

the pupils by Tropicamide 2.5% eye drops (Pharmacy of the University Hospital Aachen,

Germany), images were taken by a confocal laser scanning microscope (Heidelberg Retina

Angiograph-1, Heidelberg Engineering, Germany) in the fluorescein angiography mode.

Directly before the measurements, mice were intraperitoneally injected with 10-20 µl

fluorescein (Fluorescein Alcon® 10%, Alcon Pharma GMBH, Freiburg im Breisgau,

Germany).

2.5. Histology

At the end of the follow-up the animals were euthanized by isoflurane (Forene 100% (V/V)®,

Abott GmBH, Wiesbaden, Germany) overdosing and decapitated. Immunohistochemistry was

performed as described earlier by Mataruga et al. (2007).16

In brief, eyes were enucleated and

Page 32: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

32

RESULTS

opened by an encircling cut at the limbus. The retinae in the eyecup were immersion-fixed for

30 minutes in 4% paraformaldehyde (PA) in 0.1 M phosphate buffer (PB) at room

temperature and washed in PB several times. Tissue was incubated in 10% sucrose in PB for

1 hour, followed by 30% sucrose overnight. The retina was flat embedded and frozen in

optimal cutting temperature compound (NEG-50, Richard Allen Scientific, Thermo Fisher

Scientific, Germany). Vertical sections (20 μm thickness) were cut on a cryostat (HM 560

CryoStar, MICROM, Walldorf, Germany), and collected on Superfrost Plus slides (Menzel,

Braunschweig). Sections were pre-treated with blocking solution (5% chemiblocker

(Chemicon, Hofheim, Germany), 0.5% Triton-X100 in PB, and 0.05% NaN3) for 1 hour,

followed by incubation with primary antibodies over night, diluted in the same solution.

Sections were washed in PB and incubated in secondary antibodies diluted in 5%

Chemiblocker, 0.5% Triton-X100 in PB for 1 hour, washed in PB and coverslipped with

Aqua Polymount (Polysciences, Eppelheim, Germany). Sections were examined with a Leica

TCS confocal laser scanning microscope (Leica Microsystems, Heidelberg, Germany) with

63x/1.4 oil immersion lenses. Following primary antibodies were used: against GFAP (anti

glial fibrillary acidic protein; raised in chicken, 1:2000; Novus, Germany); against glutamine

synthetase (raised in mouse, 1:4000; BD Biosciences, Germany); against CabP (anti calbindin

28K; raised in mouse, 1:1000; Sigma); against PKC (anti protein kinase Cα; raised in rabbit,

1:4000; Santa Cruz); against Calretinin (AB1550, raised in goat, 1:3000; Millipore); against

HCN1 (RTQ-7C3, raised in rat, 1:10; F. Müller, Forschungszentrum Jülich); against recoverin

(AB5585, raised in rabbit, 1:2000; Millipore); against rhodopsin (1D4, 1:500; R.S. Molday,

Univ. of British Columbia). Secondary antibodies included: donkey anti-chicken Cy2 (1:400),

donkey anti-mouse Cy3 (1:100), donkey anti-rabbit Cy2 (1:400), donkey anti-rat Cy3 (1:500),

donkey anti-mouse Dy649 (1:500; all Dianova, Germany); donkey anti-goat Alexa647 (1:200;

Invitrogen, Germany). Peanut agglutinin (PNA, biotinylated, 1:1600; Sigma Aldrich;

Germany) was visualized using StreptavidinAlexa647 (1:100; Invitrogen, Germany).

For Hematoxylin-Eosin staining, eyes were enucleated, punctured and fixed by immersion as

described above. Eyes were dehydrated in a tissue dehydration automat (mtm, Fa. Slee,

Mainz, Germany), by incubation in a series of increasing ethanol concentrations (2 times x

70%, 2x 96%, 3x 100% for 1 hour), followed by xylene (3x 1 hour) and paraffin (4x 1 hour),

and embedded in paraffin. Sections of 5 μm thickness were cut with a microtome (R. Jung,

Page 33: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

33

RESULTS

Heidelberg, Germany), collected on slides, deparaffinized, rehydrated, and stained with

Hematoxylin and Eosin. Images were performed by a Leica DMRX microscope.

The thickness of the entire retina and of the individual retinal layers was determined in both

Hematoxylin-Eosin stainings and in immunohistochemical at six positions close to the papilla.

For each retina, thickness values obtained from these positions were averaged.

Page 34: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

34

RESULTS

5.1.4 RESULTS

ERG measurements were performed in wild type versus rd10 mice, anaesthetized with

ketamine and xylazine using goldring electrodes as active electrodes instead of contact lenses

(Figure 1A), in front of a Ganzfeld stimulator following a standardized protocol. The light-

induced electrical activity of both eyes was recorded as full-field flash ERGs as described in

the ISCEV standard protocol.[12]

Typically, the ERG waveforms consist of an early negative wave (a-wave; primary light

response in photoreceptors), followed by a large positive deflection (b-wave; dominated by

the activity of ON-bipolar cells and Müller cells). Riding on top of the b-wave are the

oscillatory potentials that probably involve inner retinal circuitry.

In wild type the amplitudes of a- and b-wave increased with light intensity and with age,

reaching their maximal levels at the age of 12 weeks (Figure 1B, 1C). In rd10 mice,

amplitudes increased only to the age of 3 weeks. In elder rd10 mice the waves of the ERG

were completely abolished (Figure 1B, 1C).

Retinal thickness was measured and the fundus of the eye was observed in vivo by Optical

Coherence Tomography (OCT) (Figure 2). For each retina, thickness was measured at six

locations close to the optic disc (examples marked by red arrows in Figure 2B, column 1).

While in wild type mice a decrease of thickness between the second and third week of age,

followed by a relatively constant retinal thickness (Figure 2, column 3) was observed, in rd10

mice a significant loss of retinal thickness was obvious (Figure 2, column 1 and 2). The outer

nuclear layer (ONL, marked by red bars in Figure 2C-F, column 1) was visible as a thin line

up to 9 weeks. Retinal separation was only observed in one 9 weeks old mouse - locally

determined directly above the optic disc - and in one 24 weeks old mouse near to the papilla

(Figure 2G, first and second column). In all other animals of each age group (n=3), no

separation was found throughout the whole retina. In Figure 2I the averaged thickness values

of the retina of wild type (Figure 2I1) and rd10 mouse (Figure 2I2) were plotted against the

age in weeks. The loss of thickness in the rd10 mice is mainly due to the thinning of the ONL

(Figure 2I3), while the inner retinal layers stayed nearly constant (Figure 2I4). The ONL

reduced its thickness up to the age of 9 weeks to 10.2% (thickness at 3 weeks: mean of 102.6

Page 35: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

35

RESULTS

µm; thickness at 9 weeks: mean of 10.44 µm; n=3 for each age group). The ONL is

completely deleted at the age of 12 weeks.

The retinae of the two strains were also examined using fluorescein angiography (Figure 3).

No significant differences were observed.

Results from the above reported non-invasive tests were compared to data obtained with

histological techniques. For this evaluation retinal areas directly near to the optic disc were

chosen, comparable to the region used for thickness measurements in the OCT (Figure 2). In

contrast to the wild type mice, in most of the rd10 mice elder than 3 weeks we observed

artificial retinal separation, probably based on the combination of photoreceptor degeneration

and the preparation technique (Figure 4).

In the OCT measurements, the wild type retina was approximately 200 ± 10 µm thick at the

regions measured, which agreed well with measurements taken in histology with a thickness

of approximately 180 to 200 µm at the corresponding position. The preparation and

histological workup of the eyes may account for the thickness difference of about 10%.

As described with the OCT, the retinal thickness of the rd10 mice decreased with the age in

HE stains (Figure 4). In 5, 7 and 9 weeks old rd10 mice, the ONL was reduced to one row of

photoreceptor somata. To further investigate degenerative changes in the rd10 mouse retina

and to identify cellular changes, immunohistochemical techniques were employed using

specific antibodies and confocal laser scanning microscopy (Figure 5). In 7 weeks old wild

type mice 10 to 12 layers of somata were visible in the ONL (Figure 5A). In accordance with

our findings in the HE staining, in the retina of 7 weeks old rd10 mice the ONL was reduced

to one row of somata (Figure 5B staining 1, green), while the inner retina seemed to be

unaffected (Figure 5B).

In Figure 5, different cell populations in the degenerated retina are visualized using three

stainings with different combinations of antibodies. The results are consistent with those

obtained by other methods: photoreceptor somata and outer segments degenerated (staining 1)

while inner retinal cells like bipolar cells, horizontal cells and amacrine cells survived

(staining 2). In wild type retina, GFAP expression was only found in astrocytes while Müller

cells were only positive for glutamine synthetase. In rd10 mice Müller cells were also positive

for GFAP, indicating that they had become reactive (Figure 5B, staining 3, green) during the

retinal degeneration process.[13]

Page 36: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

36

RESULTS

5.1.5 DISCUSSION

Animal models of retinal degenerations and dystrophies, like the rd10 mouse, are important to

study the underlying disease mechanisms and also to establish possible treatment

approaches.[3-9] No reduction of welfare of these animals over weeks was observed, although

a loss of eyesight developed after the age of three weeks. Many studies on retinal

degeneration rely on the combination of data obtained from large populations of experimental

animals, sacrificed at different stages of the degeneration process. Recently, non-invasive

tools like ERG, sd-OCT and fluorescein angiography have become promising methods that

might allow following retinal degeneration in individual animals. In the present study, we

show that the combination of these methods allows to precisely describe the degeneration of

the retina in vivo during the course of the degenerative process.

The data, we present here obtained by non-invasive methods, are in accordance with findings

of other groups.[8,11] The values of a- and b-wave in the ERG were comparable to the results

of other research groups.[8,9,11] Angiographic findings in the vasculature revealed no

significant differences between the wild type and rd10 mice.

Most importantly, our OCT scans proved most useful for quantitative analysis. The thickness

of the ONL or of the entire retina measured in vivo by OCT scans was very well comparable

to the thickness values obtained from HE staining or immunohistochemical analysis.

Pennesi et al. [14] found a separation between the outer retina and the pigment epithelium

(neurosensory detachment) in the retina of rd10 mice at the age of 64 days. In our series of

OCT scans, we were not able to confirm this finding. In vivo, we found neurosensory

detachment only locally in two animals. However, after OCT, when the eyes were prepared

for histology, separation was commonly observed probably induced during the histological

work-up. The difference between our results and those of Pennesi et al. [14] might be

explained by differences in the type of OCT machines. Alternatively, our results may indicate

that, although separation is not always manifest in vivo, it can be more easily induced in the

rd10 mouse eye than in wild type eyes.

We employed two histological methods that complemented one another. For the HE-staining

the whole eye was sectioned, which enabled us to also evaluate other parts than the neural

retina. As a disadvantage, sectioning of the entire eye is more prone to artifacts like retinal

separation that we observed especially in elder rd10 mice. Immunohistochemistry provides

Page 37: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

37

RESULTS

the advantage that the major neuronal and glial cell classes of the retina can be visualized in

much detail using cell type specific antibodies. Our immunohistochemical results confirmed

the early degeneration of rod and cone photoreceptors, the reactivity of Müller cells, as well

as neuronal remodeling processes in certain types of bipolar and horizontal cells.[15]

5.1.6 CONCLUSIONS

In summary, our data indicate that non-invasive techniques such as sd-OCT scanning of the

retina in rodents are powerful tools to monitor the course of retinal degeneration with respect

to morphometric analyses and at the same time reduce the number of animals sacrificed for

histological techniques. Quantitative anatomic parameters can be extracted from sd-OCT

scans and are in good accordance with morphometric data from histological work-up.

However, single cells and their involvement in the degenerative mechanisms can only be

identified by immunohistochemical techniques using confocal fluorescence imaging

technology.

Page 38: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

38

RESULTS

5.1.7 REFERENCES

1. E.L. Berson, “Retinitis pigmentosa: The Friedenwald Lecture”, Invest Ophthalmol Vis Sci.,

vol. 34, pp.1659-1676, 1993.

2. A. Tsubura, K. Yoshizawa, M. Kuwata, N. Uehara, “Animal models for retinitis

pigmentosa induced by MNU; disease progression, mechanisms and therapeutic trials”,

Histol Histopathol, vol. 25, pp. 933-944, 2010.

3. M.N. Delyfer, V. Forster, N. Neveux, S. Picaud, T. Léveillard, J.A. Sahel, “Evidence for

glutamate-mediated excitotoxic mechanisms during photoreceptor degeneration in the

rd1 mouse retina”, Mol Vis, vol. 11, pp. 688-696, 2005.

4. A.W. Hart, L. McKie, J.E. Morgan et al., “Genotype-phenotype correlation of mouse pde6b

mutations”, Invest Ophthalmol Vis Sci, vol. 46, pp. 3443-3450, 2005.

5. R. Gibson, E.L. Fletcher, A.J. Vingrys, Y. Zhu, K.A. Vessey, M. Kalloniatis, “Functional

and neurochemical development in the normal and degenerating mouse retina”, J Comp

Neurol, vol. 521, pp. 1251-1267, 2013.

6. C. Gargini, E. Terzibasi, F. Mazzoni, E. Strettoi, “Retinal organization in the retinal

degeneration 10 (rd10) mutant mouse: a morphological and ERG study”, J Comp

Neurol, vol. 500, pp. 222-238, 2007.

7. M.A. Chrenek, N. Dalal, C. Gardner et al., “Analysis of the RPE sheet in the rd10 retinal

degeneration model”, Adv Exp Med Biol, vol. 723, pp. 641-647, 2012.

8. B. Chang, N.L. Hawes, R.E. Hurd, M.T. Davisson, S. Nusinowitz, J.R. Heckenlively,

“Retinal degeneration mutants in the mouse”, Vision Res, vol. 42, pp. 517-525, 2002.

9. B. Chang, N.L. Hawes, M.T. Pardue et al., “Two mouse retinal degenerations caused by

missence mutations in the beta-subunit of rod cGMP phosphodiesterase gene”, Vision

Res, vol. 47, pp. 624-633, 2007.

10. D. Hicks, J. Sahel, “The Implications of Rod-Dependent Cone Survival for Basic and

Clinical Research”, Invest Ophthalmol Vis Sci, vol. 40, pp. 3071-3074, 1999.

11. S. Sugimoto, M. Imawaka, R. Imai, K. Kanamaru, T. Ito, S. Sasaki, T. Ando, T. Saijo, T.

Sato, “A procedure for electroretinogram (ERG) recording in mice - effect of

monoiodoacetic acid on the ERG in pigmented mice”, J Toxicol Sci, vol. 2, pp. 315-

325, 1997.

12. M.F. Marmor, A.B. Fulton, G.E. Holder, Y. Miyake, M. Brigell, M. Bach, “ISCEV

Standard for full-field clinical electroretinography (2008 update)”, Doc Ophthalmol,

vol. 118, pp. 69-77, 2009.

Page 39: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

39

RESULTS

13. A.J. Eisenfeld, A.H. Bund-Milam, P.V. Sarthy, “Müller cell expression of glial fibrillary

acidic protein after genetic and experimental photoreceptor degeneration in the rat

retina”, Invest Ophthalmol Vis Sci; vol. 25, pp. 1321-1328, 1984.

14. M.E. Pennesi, K.V. Michaels, S.S. Magee, A. Maricle, S.P. Davin, A.G. Garg, M.J. Gale,

D.C. Tu, Y. Wen, L.R. Erker, P.J. Francis, “Long-term characterization of retinal

degeneration in rd1 and rd10 mice using spectral domain optical coherence

tomography”, Invest Ophthalmol Vis Sci; vol. 53, pp. 4644-4656, 2012.

15. M.J. Phillips, D.C. Otteson, D.M. Sherry, “Progression of neuronal and synaptic

remodeling in the rd10 mouse model of retinitis pigmentosa”, J Comp Neurol, vol.

518, pp. 2071-2089, 2010.

16. A. Mataruga, E. Kremmer, F. Müller, “Type 3a and type 3b OFF cone bipolar cells

provide for the alternative rod pathway in the mouse retina”, J Comp Neurol, vol. 502,

pp. 1123-1137, 2007.

Page 40: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

40

RESULTS

5.1.8 FIGURES

Figure 1 Electroretinogram recordings in wild type and rd10 mice. (A) Left: Mice were

fixed in front of a Ganzfeld stimulator in a standardized position. Right: Goldring electrodes

were used on both eyes as active electrodes. A goldring electrode in the mouth served as

reference and a subcutaneous needle electrode in the lumbar region as ground electrode. (B)

Examples of ERGs of the left eye of a wild type (left) and rd10 mice (right) at different ages

(2, 3, 5 and 9 weeks). While in wild type the amplitudes of a- and b-waves increased with age,

in rd10 mice an ERG was only recordable untill the age of 3 weeks. (C) Amplitudes of the

ERG’s a- (left panel) and b-wave (right panel) in wild type mice (black bars) and rd10 mice

(grey bars) plotted with respect to the stimulus intensity (first row: 0.3 cds/m2, second row: 1

cds/m2 and third row: 3 cds/m

2) and to age given on the abscissa. Vertical bars indicate the

mean + standard deviation (n=3 per age group).

Page 41: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

41

RESULTS

Figure 2 Sd-OCT of wild type and rd10 mice. (A) Set-up. The mouse is positioned in front

of the magnifying lens of the OCT on the chin rest of an equipment usually used for

application in humans. (B) - (H) OCT images of wild type and rd10 mice at different ages.

All images are taken at the height of the papilla and displayed with the pigment epithelium to

the top. Images of rd10 mice are illustrated in two ways the first (black on white) and second

column (white on black). Red arrows in (B) (first column) demonstrate the evaluation of

thickness in the height of the papilla by six averaged cuts through the retina. Red bars in (C) -

(F) (first column) mark the outer nuclear layer (ONL). In (G) (first and second column) red

stars indicate a separation of the retina from the pigment epithelium. Separation was only

observed in vivo in one animal of nine weeks and one of 24 weeks of age (G), but not at the

age of 48 weeks. (I) Evaluation of retinal thickness in wild type and rd10 mouse in the OCT.

Values obtained from six cuts through the retina at the height of the papilla were averaged and

Page 42: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

42

RESULTS

plotted against the age (each age group: n=3). Thickness of the whole retina is illustrated in

(I1) (= wild type) and (I2) (= rd10 mouse), thickness of ONL (outer nuclear layer, somata) +

inner and outer segments of the photoreceptors (IS+OS) in (I3). In (I4) retinal thickness

except photoreceptor layers are illustrated (GC=ganglion cell layer, IPL=inner plexiform

layer, INL=inner nuclear layer and OPL=outer plexiform layer). Values represent mean ± SD

((I1) (wild type) for animals of 2 weeks: ****p<0.0001 vs. animals elder than 5 weeks; (I2)

(rd10) for animals of 2 and 3 weeks ****p<0.0001 vs. animals elder than 5 weeks; (I3) (rd10)

for animals of 2 weeks ****p<0.0001 vs. animals elder than 5 weeks; in all Figures: one-way

ANOVA with Bonferroni’s post hoc test).

Figure 3 Fluorescein angiography in wild type versus rd10 mice. In 7 and 9 weeks old

mice no significant differences in vasculature could be observed after intraperitoneal

application of fluorescein.

Page 43: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

43

RESULTS

Figure 4 Histological work up. HE stained retina. Pigment epithelium and photoreceptors

are displayed to the top. (A) Area of evaluation. The papilla is cut directly through the

median. (B) Rd10 mouse retina at the age of five weeks (left) in contrast to wild type mouse

(right). In rd10 thickness of the ONL (yellow bars) is reduced to two layers of somata and the

retina separates from the pigment epithelium (probably induced during to the preparation

process). In wild type outer segments of the photoreceptors are in contact to the pigment

epithelium (same age; ONL= outer nuclear layer, INL= inner nuclear layer, IPL= inner

plexiform layer) (C) HE-Staining of the retina of wild type (first row) and rd10 mice (second

row) in the age of 2, 5, 9, 24 and 48 weeks. While in wild type no thickness differences are

visible, in rd10 the thickness decreased with age.

Page 44: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

44

RESULTS

Figure 5 Immunohistochemical staining wild type (A) and rd10 retina (B). Staining 1:

Recoverin (all photoreceptors and type 2 bipolar cells) in green; rhodopsin (rod outer

segments, blue); HCN1 (processes in IPL, red). Staining 2: Horizontal cells (anti 28kDa

calcium-binding protein, red), rod bipolar cells (PKC, green), amacrine cells (calretinin, blue).

In rd10 retina, horizontal cells and bipolar cells lose their dendritic processes. Staining 3:

Cone outer and inner segment (PEA, blue), Müller cells (anti glutamine synthetase, red) and

astrocytes (anti GFAP, green). In rd10, Müller cell processes are also GFAP-positive. The

ONL in the rd10 mouse (B) is considerably reduced to only one row of somata.

Page 45: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

45

RESULTS

5.2 STUDY II

THE EFFECTS OF IODOACETIC ACID

ON THE MOUSE RETINA

Sarah Rösch1,2

, Sandra Johnen1, Babac Mazinani

1, Frank Müller

3, Christiane Pfarrer

2,

Peter Walter1

4. Department of Ophthalmology, RWTH Aachen University, Pauwelsstr. 30, 52074

Aachen, Germany

5. Department of Anatomy, University of Veterinary Medicine, Bischofsholer Damm 15,

30173 Hannover, Germany

6. Institute of Complex Systems, Cellular Biophysics, ICS-4, Forschungszentrum Jülich

GmbH, 52428 Jülich, Germany

SUBMITTED/ UNDER REVIEW

This work is part of a doctoral thesis at the University of Veterinary medicine Hannover

(October 2013).

Corresponding Author: Peter Walter, Department of Ophthalmology, RWTH Aachen

University, Pauwelsstr. 30, 52074 Aachen, Tel +49-241-8088191, fax +49-241-8082408, mail

to [email protected]

This work was supported by the DFG grants WA 1472/6-1 to PW and MU 3036/3-1 to FM.

Page 46: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

46

RESULTS

5.2.1 ABSTRACT

Background: To characterize the effects of intravitreal injections of iodoacetic acid (IAA) in

comparison to its systemic application as a measure to induce unilateral photoreceptor

degeneration.

Methods: Seven weeks old C57BL/6J mice received either intravitreal injections of IAA or

systemic treatment (intraperitoneal vs. intravenous) and were observed in the following five

weeks using ERG, OCT, and histology.

Results: Systemic treatment with IAA showed high systemic toxic effects and a high

mortality in contrast to the intravitreal injection. Intraperitoneal application had no effect on

the retina. Intravenous application of 2 x 30 mg/kg BW IAA (time distance 3.5 hours)

resulted in an extinction of the ERG and a thinning of the retina, in particular of the outer

nuclear layer (ONL) indicating a photoreceptor degeneration. Animals receiving intravitreal

injections developed cataracts already in low concentrations up to 100% at 0.25 mg/kg BW.

Higher intravitreal IAA doses lead to extinguished ERGs. In histology a thinning of the entire

retina was observed that was most prominent in the inner part of the retina.

Conclusions: In contrast to intraperitoneal, intravenous application of IAA leads to a

selective photoreceptor degeneration. After intravitreal injection dense cataracts are already

observed at concentrations lower than those needed to induce changes in the ERG. ERG

results must be interpreted carefully. A thinning of all retinal layers rather than a specific

outer retinal degeneration was observed upon intravitreal injection. IAA is not a useful model

to study outer retinal degeneration in mice.

Page 47: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

47

RESULTS

5.2.2 INTRODUCTION

Animal models for retinal degenerations are important tools to explore experimental therapies

for currently untreatable conditions. In rodents, genetic models are available resembling

photoreceptor degenerations such as observed in Retinitis pigmentosa (RP). In mice, for

example the rd1 and the rd10 mutants, carrying mutations in the gene encoding the ß-subunit

of rod cGMP phosphodiesterase, were described and widely used.1-9

In these mice,

comparable to RP, a progressive loss of photoreceptors - first rods, then cones - with age was

observed, concomitant with a pronounced reduction in the thickness of the outer nuclear layer

(ONL), while the thickness of the inner retinal layers remained unaffected.10,11

As a rat model,

the RCS (Royal College of Surgeons) rat was described and used.12-14

Experimental

treatments, e.g. the application of growth factors, were evaluated in such animal models

already years ago.15

To evaluate experimental surgical approaches for the treatment of

inherited retinal degeneration, models of animal species with larger eyes are desirable.

Nishida et al. used photodynamic therapy to induce an outer retinal degeneration in rabbits to

test the efficacy of suprachoroidal electrical stimulation.16

Recently, a genetic model was

described in rabbits17-19

, however, maintaining such a transgenic colony may be very cost

intensive and will also require other resources, possibly not easily available. Beside genetic

models, several induced models for retinal degeneration have been described. Among them

are degenerations induced by light20

and by systemically applied pharmaceuticals such as

iodoacetic acid (IAA)22-37

or N-methyl-N-nitrosourea (MNU)21

. It was described that IAA

leads to an elimination of photoreceptors while neurons of the inner retinal layers remain

unaffected.22-24

IAA covalently modifies and inhibits the glyceraldehyde 3-phosphate

dehydrogenase (GAPDH), an essential enzyme for glycolysis, which is halted.22,25

Because of

their very high metabolic rate, photoreceptors seem to be particularly sensitive for IAA, but

also other cells are influenced in their energy balance.23

Iodoacetic acid is not only reported to

irreversibly modify thiol (SH) groups as in GAPDH, it also reacts with imidazole and amino

groups of proteins,26

so that toxic side effects after systemic application are likely. The

intravenous application resulted in a degeneration of photoreceptors in mouse24

, rat26-28

,

ground squirrel29

, rabbit30-34

, pig35,36

, and monkey37

. Intravenous treatment with IAA also

causes cataracts after a latent period of one to three months30

hypothetically caused by a

blockage of the energy metabolism in the lens and inhibition of mitosis in the lens epithelium,

Page 48: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

48

RESULTS

as a result of damage to the SH-dependent enzymatic systems.33

Further, a breakdown of the

blood-aqueous barrier resulting in an plasmoid and cellular exudation into the aqueous humor

after two to five days was reported.30,33

Intraperitoneal application is rarely described but also

leads to cataract formation twenty weeks after injection.26

However, both ways of systemic

administration are affecting the general well-being of the animal.

To avoid systemic side effects combined with the advantage of having an intraindividual

control eye, we investigated the effects of intravitreal injections of iodoacetic acid in the

retina. We hypothesized that local administration of IAA by intravitreal injection will induce

photoreceptor degeneration similar to the effects obtained with systemic application, but

without affecting the fellow eye and without affecting the general health status of the animal.

Page 49: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

49

RESULTS

5.2.3 MATERIALS & METHODS

The materials and methods were performed as also described in Study I: ”Correlations

between ERG, OCT and anatomical findings in the rd10 mouse” and Study III: “Selective

photoreceptor degeneration by intravitreal injection of N-methyl-N-nitrosourea”.

All animal experiments adhered to the “Principles of laboratory animal care” (NIH

publication No. 85-23, revised 1985), the OPRR Public Health Service Policy on the Humane

Care and Use of Laboratory Animals (revised 1986) and the U.S. Animal Welfare Act, as

amended, as well as the current version of the German law on the protection of animals and

the local commission for animal welfare. The proposal of the animal experiments was

approved by the LANUV (Landesamt für Natur, Umwelt und Verbraucherschutz Nordrhein

Westfalen, Recklinghausen, Germany; file reference: 84-02.04.2011.A386).

Animals

Adult pigmented female wild type mice (C57BL/6J; 6 - 8 weeks) were maintained in an air-

conditioned environment under controlled light conditions (12:12 hours light/dark cycle) with

free access to food and water. Animals received sterile iodoacetic acid (IAA; Sigma Aldrich,

Seelze, Germany) in different concentrations systemically either as intraperitoneal injections

(8 animals, 40 - 70 mg/kg bodyweight (BW)) or as intravenous injections (12 animals, 55 - 65

mg/kg BW). A third group of animals received IAA as intravitreal injections (25 animals,

between 0.1 - 1 mg/kg BW). The sodium salt of IAA was dissolved in phosphate buffered

saline and neutralized to pH 7.4 with dilute NaOH immediately before use. The animals were

examined 5 days before, 7 days, 14 days and single animals up to five weeks after injection of

IAA to monitor the photoreceptor function and effects of the injection. Histological

examinations were made at two or five weeks after treatment.

For all experimental procedures and examinations except for intraperitoneal or intravenous

injections of IAA the animals were anaesthetized with an intraperitoneal injection of a

mixture of ketamine (70 mg/kg Ketamin® 10%, CEVA, Germany) and xylazine (10 mg/kg

Xylazin 2% Bernburg®, Medistar, Ascheberg, Germany) in 0.1 mL of saline.

Page 50: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

50

RESULTS

Systemic IAA-application

For systemic application, the neutralized IAA-solution was administered intraperitoneally or

intravenously via one of the tail veins at different concentrations. The IAA-solution was at

room temperature. Very rapid injections were avoided. The concentrations were chosen

around the value of 60 mg/kg BW - a concentration reported to effectively cause

photoreceptor degeneration in mice.24

Eight animals were treated with an intraperitoneal injection of IAA: 40 mg/kg BW (n=1), 50

mg/kg BW (n=1), 60 mg/kg BW (n=2), 65 mg/kg BW (n=1), 70 mg/kg BW (n=1), 75 mg/kg

BW (n=1), and 2 x 30 mg/kg BW in a time interval of 3.5 hours (n=1).

Twelve animals received intravenous injections of IAA: 55 mg/kg BW (n=2), 60 mg/kg BW

(n=5), 65 mg/kg BW (n=1), and 2 x 30 mg/kg BW in a time interval of 3.5 hours (n=4).

Intravitreal IAA-injection

Animals were anesthetized as described before and eyes were locally anesthetized by

proxymetacainhydrochloride 0.5% eye drops (Proparakain- POS®, Ursapharm, Saarbrücken,

Germany). Eyes were punctured by a 27 gauge steel needle (BD Microlance TM

3, Heidelberg,

Germany). Intravitreal injections were performed using a nanoliter injection system with a 3D

micromanipulator (Nanoliter 2000; World Precision Instruments, Inc. Sarasota, FL, USA).

Injection volume was fixed to 2 µL. Preliminary experiments showed that this volume could

be injected safely into the vitreous cavity without touching the lens or the retina. A standard

volume of 2 µL with different IAA-concentrations was injected intravitreally in 25 mice: 0.1

mg/kg BW (n=3), 0.15 mg/kg BW (n=5), 0.2 mg/kg BW (n=6), 0.25 mg/kg BW (n=4), 0.5

mg/kg BW (n=4), 1 mg/kg BW (n=3).

After intravitreal injections, the animals were treated with antibiotic eye ointment

(Polyspectran, Alcon Pharma GmbH, Freiburg, Germany) up to three days after application.

Control interventions

Saline was injected as a control substance in the tail vein of one mouse, in the peritoneum of

three mice, and in the vitreous cavity of one eye in three mice. Control mice received the

equivalent volume of saline as the IAA treated animals. Control animals were examined five

Page 51: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

51

RESULTS

days before, after one week, and two weeks up to four weeks after injection by recording

ERG and OCT and were finally sacrificed for histology.

In three control animals, 2 µL fluorescein solution was injected intravitreally to confirm the

correct placement of the injection cannula and the absence of leakage from the injection area.

Directly after injection images were taken by a confocal laser scanning microscope

(Heidelberg Retina Angiograph-1, Heidelberg Engineering, Germany) in the fluorescein

angiography mode to exclude side effects possibly resulting from the application technique.

Electroretinogram recordings

Electroretinogram recordings (ERG) were performed with the Reti System especially

designed for mice (Roland Consult Electrophysiological Diagnostic Systems, Brandenburg,

Germany). Animals were dark adapted for one hour. The pupils were dilated using

Tropicamide 2.5% eye drops (Pharmacy of the University Hospital Aachen, Germany) and

subsequently anaesthetized with proxymetacainhydrochloride 0.5% eye drops (Proparakain-

POS®, Ursapharm, Saarbrücken, Germany). Goldring electrodes (animal electrode 0.5mm ø

3mm Roland Consult) were placed as active electrodes on the corneal surface of both eyes.

Methylcellulose (Methocel® 2%, Omni Vision, Puchheim, Germany) was applied to keep the

mouse eye hydrated and maintain a good ionic conduction. The reference goldring electrode

was placed on the mouth mucosa and a subcutaneous silver needle electrode in the lumbar

region served as ground electrode. The fullfield-ERG was performed according to the ISCEV

standard protocol.38

Five scans to light stimulation were performed averaged at different light

intensities. A- and b-wave amplitudes and implicit times of rod and cone responses were

determined.

Spectral domain Optical Coherence Tomography

Spectral domain Optical Coherence Tomography (sd-OCT) images were obtained using the

Spectralis® OCT system (Heidelberg Engineering, Heidelberg, Germany). Retinal images

were centered on the optic disk as the main landmark and retinal thickness was determined at

six positions across the retina (Fig. 3b red arrows). For each eye these thickness values were

averaged. The thickness of the different layers of the retina was determined in the same way.

Page 52: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

52

RESULTS

The fundus of each eye was recorded by the sd-OCT system (IR reflection image, wavelength

715nm).

Macroscopic pictures of the eye appearance were taken with a Nikon D80 camera (Objective:

Nikon AF Micro Nikkor 105 mm, 2896 Pixel x 1944 Pixel x 24 Bit).

Histology

At the end of the follow-up the animals were euthanized by isoflurane (Forene 100% (V/V),

Abott GmBH, Wiesbaden, Germany) overdosing, decapitated and enucleated.

Immunohistochemistry was performed as described earlier by Mataruga et al. (2007).40

In

brief, eyes were opened by an encircling cut at the limbus. The retinae in the eyecup were

immersion-fixed for 30 min in 4% paraformaldehyde (PA) in 0.1 M phosphate buffer (PB) at

room temperature, washed in PB several times and incubated in 10% sucrose in PB for 1 h,

followed by 30% sucrose overnight. The retina was flat embedded and frozen in optimal

cutting temperature (OCT) compound (NEG-50, Richard Allen Scientific, Thermo Fisher

Scientific, Germany). Vertical sections (20 µm thickness) were cut on a cryostat (HM 560

CryoStar; MICROM; Walldorf; Germany) and collected on Superfrost Plus slides (Menzel,

Braunschweig). Sections were pre-treated with blocking solution (5% chemiblocker

(Chemicon, Hofheim, Germany), 0.5% Triton-X100 in PB, and 0.05% NaN3) for 1 hour,

followed by incubation with primary antibodies over night, diluted in the same solution.

Sections were washed in PB and incubated in secondary antibodies diluted in 5%

Chemiblocker, 0.5% Triton-X100 in PB for 1 hour, washed in PB and coverslipped with

Aqua Polymount (Polysciences, Eppelheim, Germany). Sections were examined with a Leica

TCS confocal laser scanning microscope (Leica Microsystems, Heidelberg, Germany) with

63x/1.4 oil immersion lenses. Images were processed and printed with Adobe Photoshop. For

double or triple labelling, primary antibodies were mixed and applied simultaneously. All

secondary antibodies were highly cross-absorbed and were carefully tested to exclude

reactions with the wrong primary antibody. Concentration of the antibodies, laser intensity,

and filter settings were carefully controlled and the sequential scanning mode was employed

to completely rule out cross-talk between the fluorescence detection channels. Band pass

filters of 500–530 nm for green fluorescence (Cy2), 580–650 nm for red fluorescence (Cy3),

and 680–750 nm for infrared fluorescence (Alexa647) were used. Primary antibodies were

Page 53: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

53

RESULTS

directed against following epitopes: GFAP (anti glial fibrillary acidic protein; raised in

chicken, 1:2000; Novus, Germany) and glutamine synthetase (raised in mouse, 1:4000; BD

Biosciences, Germany). Secondary antibodies included donkey anti-chicken Cy2 (1:400,

Dianova, Germany) and donkey anti-mouse Cy3 (1:100, Dianova, Germany). The lectin

Peanut agglutinin (PNA, biotinylated, 1:1600; Sigma Aldrich; Germany) was visualized using

StreptavidinAlexa647 (1:100, In vitrogen, Germany).

For Hematoxylin-Eosin staining, eyes were enucleated, punctured three times at the limbus

and fixed by immersion as described above. Eyes were dehydrated (dehydration automat,

mtm, Slee, Mainz, Germany) by incubation in a series of increasing ethanol concentrations (2

times x 70%, 2 x 96%, 3 x 100% for one hour), followed by xylene (3 times 1 hour) and

paraffin (4 times one hour), and embedded in Paraffin. Sections of 5 µm thickness were cut

with a microtome (R. Jung, Heidelberg, Germany), collected on slides, deparaffinized,

rehydrated, and stained with Hematoxylin and Eosin. Pictures were taken using a Leica

DMRX microscope.

The thickness of the entire retina and of the individual retinal layers was determined in both

immunohistochemical and in Hematoxylin-Eosin stainings at six positions close to the papilla.

For each retina, thickness values obtained from these positions were averaged.

Page 54: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

54

RESULTS

5.2.4 RESULTS

Systemic application of IAA. Intraperitoneal and intravenous application of iodoacetic acid

resulted in considerable systemic side effects in mice. Intraperitoneal application of IAA

induced mortality of 25% and weight loss within the first two days of about 10% unrelated to

the dosage given. When IAA was injected intravenously, a mortality of 60%, but only little

effect on body weight was observed. Mortality after injection of IAA in the vein was related

to dosage and death occurred within the first four hours after injection. In contrast, when IAA

was administered intravitreally, no systemic side effects or even lethal outcomes were

observed. The results are summarized in Figure 1.

To monitor the functionality of the retina, light-induced electrical activity of both eyes was

recorded as full-field flash ERGs as described in the ISCEV standard protocol.38

Typically,

the ERG waveforms consist of an early negative wave (a-wave), followed by a large positive

deflection (b-wave). Whereas at least the initial portion of the a-wave reflects the primary

light response in photoreceptors, the b-wave is dominated by the activity of ON-bipolar cells

and Müller cells. Riding on top of the b-wave are the oscillatory potentials, small wavelets

that probably involve inner retinal circuitry. Systemically administered IAA did not affect the

amplitudes of either a-wave or b-wave (Fig. 2b, 2d), except when 2 x 30 mg/kg BW IAA

were applied intravenously (Fig. 2e). In these cases a reduction of the amplitudes of the a- and

b-waves could be seen as well as a loss of the oscillatory potentials.

Following the ERG recordings, retinal thickness was measured and the fundus of the eye was

observed in vivo by optical coherence tomography (OCT). Intraperitoneal application of IAA

did not induce any quantitative changes in retinal thickness as determined from OCT scans.

For each retina, thickness was measured at six locations close to the papilla (examples marked

by red arrows in Figure 3b). IAA concentrations below 60 mg/kg BW given as intravenous

injections were also not effective in reducing retinal thickness within the follow-up. Only

when 30 mg/kg BW was injected twice within 3.5 hours, a significant reduction of retinal

thickness was seen as demonstrated in Figure 3e (red bars) and Figure 3h.

OCT displayed no effect of systemic IAA administration on the retinal thickness, except for

the animal treated with 2 x 30 mg/kg BW IAA injected in the tail vein (Figure 3). This finding

was corroborated by immunohistochemical analysis of vertical sections through the retinae of

all experimental animals stained with antibodies against glutamine synthetase to visualize

Page 55: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

55

RESULTS

Müller cells, PNA to label cone outer segments and endfeet, and anti-GFAP antibodies. Under

normal conditions, GFAP is only expressed in astrocytes. However, during retinal

degenerations, GFAP may also be observed in Müller cells, that have become reactive.39

Two

representative examples are shown in Figure 4. Only in the animal treated with the double

injection of IAA, the retinal thickness was considerably reduced due to the thinning of the

outer nuclear layer (ONL) to only 3-5 rows of photoreceptor somata (Fig. 4c), while the inner

retina seemed to be unaffected. Müller cells became reactive, as indicated by GFAP

expression (Fig. 4d). The thinning of the outer retina was reminiscent of - albeit not as

pronounced as - the photoreceptor loss observed in the retina of the rd10 mouse (Fig. 4e,

Biswas et al., in preparation).6-14

Intravitreal injection of IAA. Animals that received iodoacetic acid as intravitreal injections

showed no remarkable weight loss or other systemic side effects (see Figure 1a and 1b).

However, ocular side effects occurred in relation to the dose given. Cataract and inflammatory

reactions were most prominent. Even at the low concentration of 0.1 mg/kg BW, 20% of the

injected eyes developed cataract. With concentrations of 0.25 mg/kg BW or higher, phthisis

bulbi was frequently observed and the ERG was not recordable (Fig. 5). Due to the frequent

manifestation of cataracts and inflammatory infiltrates in the aqueous humor as verified by

HE-staining (data not shown), OCT scanning could only be performed in eyes that had

received low dosages of IAA. In those eyes where pictures could be taken, OCT recordings

were blurry and the layering of the retina seemed less clear two weeks after the injection (Fig.

6). This finding was inconsistent and in the immunohistochemical analysis the retinae showed

a normal structure and thickness (Fig. 8b). In control eyes injected with PBS, no changes in

retinal layering were observed (see Fig. 3f and 3i).

With increasing concentrations of intravitreal IAA considerable changes in the ERG were

found. ERG a- and b-wave were reduced or extinguished at concentrations of 0.25 mg/kg BW

or higher as shown in Figure 7.

In experiments with higher doses of intravitreal iodoacetic acid, histology revealed a general

thinning of the retina after two weeks (Figure 8). A specific effect of IAA on photoreceptor

survival should lead to a thinning of the outer nuclear layer, while the inner retina should

remain intact. In contrast to this, thinning was more pronounced in the inner retina (reduction

Page 56: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

56

RESULTS

up to 44% measured in immunohistochemical sections) while only small changes were

observed in the outer nuclear layer (reduction up to 16%). Glial reaction was present as

indicated by GFAP immunoreactivity in Müller cells (Fig. 8e). Concentrations of 0.5 mg/kg

BW IAA and higher led to a gross disorganization of the retina, partially with a total

destruction of the retinal layers with interindividual differences so that morphometric

measurements did not reveal any concise results (data not shown). In all experiments, the

untreated control eyes of the animals were not affected and showed neither changes in ERG

nor in OCT measurements.

Page 57: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

57

RESULTS

5.2.5 DISCUSSION

Animal models of retinal degenerations and dystrophies are important to study the underlying

disease mechanisms and also to establish possible treatment approaches. For retinal

dystrophies such as Retinitis pigmentosa (RP) several rodent models are established. Beside

these genetic models photoreceptor degeneration may also be induced pharmacologically. The

non-genetic models may have the advantage that they can be induced in a variety of animals

with larger eyes possibly more suitable to work on surgical methods for the treatment of

retinal dystrophies. A number of publications demonstrated that iodoacetic acid (IAA) may

lead to a blindness causing photoreceptor degeneration when administered intravenously. The

resulting histological picture is very similar to that presented in human RP.23

However, the

systemic application of IAA not only leads to bilateral disease but also to considerable

systemic side effects. Therefore, we hypothesized that IAA given intravitreally may induce

unilateral photoreceptor degeneration in mice without causing systemic side effects. The

systemic application of IAA either given as an intraperitoneal or as an intravenous injection

served as positive control.

In animals receiving systemic application, significant side effects occurred. The well-being of

the animals was strongly reduced for some hours after intravenous administration and for

some days after intraperitoneal application of IAA. Upon intravenous application, the

mortality rate in mouse was 60%, which is higher than in rat with 50%28

or in rabbit with

20%32

. Of the 12 intravenously injected animals of our study, only five survived- two with 55

mg/kg BW, two with 60 mg/kg BW and one animal which was treated by two shots of 30

mg/kg BW separated by 3.5 hours. The concentrations being used in our experiments were

derived from work of Sugimoto et al.24

Splitting the 60 mg/kg BW into two dosages given

within 3.5 hours as described in rat by Graymore et al.28

did not lower the mortality in the

mouse. But similar to their finding, also in mice 60 mg/kg BW IAA was found to more

effectively reduce photoreceptor density when administered as a divided dose. Probably the

two injection paradigm increased the pharmaceutical effect by maintaining a critical

concentration of IAA for a sufficient length of time28

. In our experiments, animals were

sacrificed two or five weeks after the injection. By that time in the two shot paradigm, the

ONL was reduced to 3 – 5 layers of somata. Detailed analysis using markers for cones (e.g.

Fig. 4c, other markers not shown) and rods (data not shown) indicate that the surviving

Page 58: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

58

RESULTS

photoreceptor population comprised both rods and cones and that outer segments were

present. As our systemic application of IAA only served as positive controls, we did not

attempt to either monitor the time course of degeneration or to quantify whether the two cone

photoreceptor types are affected to different extent by IAA. The incomplete loss of

photoreceptors after two weeks is in agreement with histological examinations in IAA treated

rabbits (some photoreceptors survived up to four months)34

and pigs (up to 12 weeks with a

progressive loss of rods in time)35

. Interestingly, the expected effects of IAA on the retina

were not seen after intraperitoneal injection suggesting a first pass effect in the liver. The

retina remained unaffected. We did not observe cataractous changes in mice receiving

systemic application of IAA, probably because under these experimental conditions

development of cataracts was reported to be slow.23,28

Intravitreal injections in mice are difficult because the lens occupies most of the lumen of the

eye. The vitreous space is small and injections are difficult to place without touching the lens

or the retina. Therefore, we used a 3D micromanipulator and nanoliter injection system for the

injections. Injections of PBS, fluorescein and other substances carried out as negative controls

demonstrated that the injection procedure was safe and did not induce any changes in the lens

or the retina. In contrast to the systemic application the welfare of the animals was only

minimally affected upon intravitreal injections of IAA. We only noticed a slight weight loss

similar to that observed normally after an anesthetic intervention. However, the intravitreal

injection of IAA induced severe side effects, like cataract and inflammatory responses, on the

eye similar to those described after systemic application in longer experimental trials. Most

likely, the cataractous changes were not the result of a lens injury during the injection because

they appeared at about five to seven days after injection and they were not observed in control

animals receiving injections other than iodoacetic acid. Inflammatory responses with

plasmoid, fibrinous, and cellular exudation into the aqueous humor were also described by

Cibis et al. after intravenous application of IAA.23

All the described effects of iodoacetic acid

occurred with interindividual differences. This variable effect of the toxin was already

reported after intravenous application in the rabbit.23

Finally and most importantly, in contrast to the expected effect, no selective thinning of the

outer retina was observed upon intravitreal injection of IAA. Rather an overall thinning of the

retina was detected, with a tendency towards a more pronounced reduction in the inner retina.

Page 59: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

59

RESULTS

Our results clearly indicate that IAA given intravitreally is not suitable to induce

photoreceptor degeneration to model disease processes as seen in RP or similar conditions.

Because of the cataract formation, retinal changes could not be monitored by OCT recordings

and also ERG recordings may be affected. Based on these results, we would not recommend

using IAA injections in the vitreous of mice or larger animals as a pharmaceutical model to

specifically induce degeneration of photoreceptors.

5.2.6 ACKNOWLEDGEMENT

The authors thank Christoph Aretzweiler for the excellent support in preparation and staining

of the eyes. This work was supported by the DFG grants WA 1472/6-1 to PW and MU

3036/3-1 to FM.

Page 60: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

60

RESULTS

5.2.7 REFERENCES

1. Delyfer MN, Forster V, Neveux N, Picaud S, Léveillard T, Sahel JA. (2005) Evidence for

glutamate-mediated excitotoxic mechanisms during photoreceptor degeneration in the

rd1 mouse retina. Mol Vis;11:688-696.

2. Hart AW, McKie L, Morgan JE, Gautier P, West K, Jackson IJ, Cross SH. (2005)

Genotype-phenotype correlation of mouse pde6b mutations. Invest Ophthalmol Vis

Sci;46:3443-3450.

3. Ahuja S, Ahuja-Jensen P, Johnson LE, Caffé AR, Abrahamson M, Ekström PA, van Veen

T. (2008) Rd1 Mouse retina shows an imbalance in the activity of cysteine protease

cathepsins and their endogenous inhibitor cystatin C. Invest Ophthalmol Vis

Sci;49:1089-1096.

4. Gibson R, Fletcher EL, Vingrys AJ, Zhu Y, Vessey KA, Kalloniatis M. (2013) Functional

and neurochemical development in the normal and degenerating mouse retina. J Comp

Neurol;521:1251-1267.

5. Gargini C, Terzibasi E, Mazzoni F, Strettoi E. (2007) Retinal organization in the retinal

degeneration 10 (rd10) mutant mouse: a morphological and ERG study. J Comp

Neurol;500:222-238.

6. Chen M, Wang K, Lin B. (2012) Development and degeneration of cone bipolar cells are

independent of cone photoreceptors in a mouse model of retinitis pigmentosa. PLoS

ONE;7:e44036.

7. Samardzija M, Wariwoda H, Imsand C, Huber P, Heynen SR, Gubler A, Grimm C. (2012)

Activation of survival pathways in the degenerating retina of rd10 mice. Exp Eye

Res;99:17-26.

8. Chrenek MA, Dalal N, Gardner C, Grossniklaus H, Jiang Y, Boatright JH, Nickerson JM.

(2012) Analysis of the RPE sheet in the rd10 retinal degeneration model. Adv Exp Med

Biol;723:641-647.

9. Cang B, Hawes NL, Hurd RE, Davisson MT, Nusinowitz S, Heckenlively JR. (2002)

Retinal degeneration mutants in the mouse. Vision Res;42:517-525.

Page 61: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

61

RESULTS

10. Chang B, Hawes NL, Pardue MT, German AM, Hurd RE, Davisson MT, Nusinowitz S,

Rengarajan K, Boyd AP, Sidney SS, Phillips MJ, Stewart RE, Chaudhury R, Nickerson

JM, Heckenlively JR, Boatright JH. (2007) Two mouse retinal degenerations caused by

missence mutations in the beta-subunit of rod cGMP phosphodiesterase gene. Vision

Res;47:624-633.

11. Pang JJ, Boye SL, Kumar A, Dinculescu A, Deng W, Li J, Li Q, Rani A, Foster TC,

Chang B, Hawes NL, Boatright JH, Hauswirth WW. (2008) AAV-mediated gene

therapy for retinal degeneration in the rd10 mouse containing a recessive PDEbeta

mutation. Invest Ophthalmol Vis Sci;49:4278-4283.

12. Hess HH, Newsome DA, Knapka JJ, Westney GE. (1982) Slitlamp assessment of age of

onset and incidence of cataracts in pink-eyed, tan-hooded retinal dystrophic rats. Curr

Eye Res;2:265-269.

13. Matuk Y. (1984) Retinitis pigmentosa and retinal degeneration in animals: a review. Can

J Biochem Cell Biol;62:535-546.

14. Fletcher EL, Kalloniatis M. (1997) Neurochemical development of the degenerating rat

retina. J Comp Neurol;388:1-22.

15. Baid R, Upadhyay AK, Shinohara T, Kompella UB. (2013) Biosynthesis,

Characterization, and Efficacy in Retinal Degenerative Diseases of Lens Epithelium-

derived Growth Factor Fragment (LEDGF1-326), a Novel Therapeutic Protein. J Biol

Chem;288:17372-17383.

16. Nishida K, Kamei M, Kondo M, Sakaguchi H, Suzuki M, Fujikado T, Tano Y. (2010)

Efficacy of suprachoroidal-transretinal stimulation in a rabbit model of retinal

degeneration. Invest Ophthalmol Vis Sci;51:2263-2268.

17. Jones BW, Kondo M, Terasaki H, Watt CB, Rapp K, Anderson J, Lin Y, Shaw MV, Yang

JH, Marc RE. (2011) Retinal remodeling in the Tg P347L rabbit, a large-eye model of

retinal degeneration. J Comp Neurol;519:2713-2733.

18. Muraoka Y, Ikeda HO, Nakano N, Hangai M, Toda Y, Okamoto-Furuta K, Kohda H,

Kondo M, Terasaki H, Kakizuka A, Yoshimura N. (2012) Real-time imaging of rabbit

retina with retinal degeneration by using spectral-domain optical coherence

tomography. PLoS ONE;7:e36135.

Page 62: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

62

RESULTS

19. Hirota R, Kondo M, Ueno S, Sakai T, Koyasu T, Terasaki H. (2012) Photoreceptor and

post-photoreceptoral contributions to photopic ERG a-wave in rhodopsin P347L

transgenic rabbits. Invest Ophthalmol Vis Sci;53:1467-1472.

20. Harada T, Harada C, Sekiguchi M, Wada K. Light-induced retinal degeneration

suppresses developmental progression of flip-to-flop alternative splicing in GluR1. J

Neurosci. 1998;18:3336-3343.

21. Tsubura A, Yoshizawa K, Kuwata M, Uehara N. (2010) Animal models for retinitis

pigmentosa induced by MNU; disease progression, mechanisms and therapeutic trials.

Histol Histopathol;25:933-944.

22. Liang L, Katagiri Y, Franco LM, Yamauchi Y, Enzmann V, Kaplan HJ, Sandell JH.

(2008) Long-term cellular and regional specificity of the photoreceptor toxin,

iodoacetic acid (IAA), in the rabbit retina. Vis Neurosci;25:167-177.

23. Noell WK. (1953) Experimentally induced toxic effects on structure and function of

visual cells and pigment epithelium. Am J Ophthalmol;36:103-116.

24. Sugimoto S, Imawaka M, Imai R, Kanamaru K, Ito T, Sasaki S, Ando T, Saijo T, Sato T.

(1997) A procedure for electroretinogram (ERG) recording in mice – effect of

monoiodoacetic acid on the ERG in pigmented mice. J Toxicol Sci;2:315-325.

25. Winkler BS, Sauer MW, Starnes CA. (2003) Modulation of the Pasteur effect in retinal

cells: implications for understanding compensatory metabolic mechanisms. Exp Eye

Res;76:715-723.

26. Matsumoto M, Morita T, Hirayama S, Uga S, Shimizu K. (2002) Morphological analysis

of Iodoacetic acid-induced cataract in the rat. Ophthalmic Res;34:119-127.

27. Graymore C, Tansley K. (1959) Iodoacetate poisoning of the rat retina. I. Production of

retinal degeneration. Br J Ophthalmol;43,177-185.

28. Graymore C, Tansley K. (1959) Iodoacetate poisoning of the rat retina. II. Glycolysis in

the poisoned retina. Br J Ophthalmol;43:486-493.

29. Farber DB, Souza DW, Chase DG. (1983) Cone visual cell degeneration in ground

squirrel retina: disruption of morphology and cyclic nucleotide metabolism by

Iodoacetic acid. Invest Ophthalmol Vis Sci;24:1236-1249.

30. Cibis PA, Constant M, Pribyl A, Becker B. (1957) Ocular lesions produced by

Iodoacetate. AMA Arch Ophthalmol;57:508-519.

Page 63: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

63

RESULTS

31. Lasansky A, De Robertis E. (1959) Submicroscopic changes in visual cells of the rabbit

induced by iodoacetate. J Biophys Biochem Cytol;5:245-250.

32. Orzalesi N, Calabria GA, Grignolo A. (1970) Experimental degeneration of the rabbit

retina induced by iodoacetic acid. A study of the ultrastructure, the rhodopsin cycle and

the uptake of 14C-labelled iodoacetic acid. Exp Eye Res;9:246-253.

33. Tieri O, Vecchione L. (1963) Monolateral cataract provoked with iodoacetic acid. Acta

Ophthalmol (Copenh);41:205-212.

34. Yamauchi Y, Agawa T, Tsukahara R, Kimura K, Yamakawa N, Miura M, Goto H. (2011)

Correlation between high-resolution optical coherence tomography (OCT) images and

histopathology in an iodoacetic acid-induced model of retinal degeneration in rabbits.

Br J Ophthalmol;95:1157-1160.

35. Wang W, Fernandez de Castro J, Vukmanic E, Zhou L, Emery D, Demarco PJ, Kaplan

HJ, Dean DC. (2011) Selective rod degeneration and partial cone inactivation

characterize an iodoacetic acid model of Swine retinal degeneration. Invest Ophthalmol

Vis Sci;52:7917-7923.

36. Scott PA, Kaplan HJ, Sandell JH. (2011) Anatomical evidence of photoreceptor

degeneration induced by iodoacetic acid in the porcine eye. Exp Eye Res;93:513-527.

37. Noell WK. (1952) The impairment of visual cell structure by iodoacetate. J Cell

Physiol;40:25-55.

38. Marmor MF, Fulton AB, Holder GE, Miyake Y, Brigell M, Bach M. (2009) ISCEV

Standard for full-field clinical electroretinography (2008 update). Doc

Ophthalmol;118:69-77.

39. Eisenfeld AJ, Bund-Milam AH, Sarthy PV. (1984) Müller cell expression of glial

fibrillary acidic protein after genetic and experimental photoreceptor degeneration in

the rat retina. Invest Ophthalmol Vis Sci;25(11):1321-1328.

40. Mataruga A, Kremmer E, Müller F. (2007) Type 3a and type 3b OFF cone bipolar cells

provide for the alternative rod pathway in the mouse retina. J Comp Neurol;502:

1123-1137.

Page 64: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

64

RESULTS

5.2.8 FIGURES

Figure 1 Systemic side effects of IAA application. a Loss of body weight as an indicator of

general well-being after application of IAA given in different routes. ****p<0.0001 vs.

intraperitoneal (i.p.) application, one-way ANOVA with Bonferroni’s post hoc test.

(intravenous (i.v.): n=12; i.p.: n=8; intravitreal (IVI): n=25) b Body weight reduction after i.p.

IAA injection. For each concentration n=1; control animals (n=3 i.p.) were injected with the

same volume of PBS. c Mortality within the first two days after IAA injection. (i.v.: n=12;

i.p.: n=8; IVI: n=25). d Mortality after i.v. injection of IAA and concentration of IAA given

(control: n=1; 55 mg/kg BW: n=2; 60 mg/kg BW: n=5; 2 x 30 mg/kg BW: n=4; 65 mg/kg

BW: n=1).

Page 65: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

65

RESULTS

Figure 2 ERG after systemic IAA application. a – e Examples of full-field ERG recordings

from left eyes of mice before (black line) and one week after (grey line) systemic application

of IAA or PBS [Scotopic 3 cds/m2, 0.067 Hz]. Only in e an extinction of the ERG curve one

week after IAA application could be observed. a PBS i.p. b IAA 65 mg/kg BW i.p. c PBS i.v.

d IAA 60 mg/kg BW i.v. e IAA 2 x 30 mg/kg BW i.v. f, g Amplitude of a-wave (f) and b-

wave (g) before and two weeks after IAA application at a light intensity of 3 cds/m2 in the

scotopic ERG. Mean + SD. PBS injections served as controls.

Page 66: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

66

RESULTS

Figure 3 Thickness evaluation in the OCT. Retinal images and thickness obtained by sd-

OCT measurements. Left panel. All images are taken at the height of the papilla and

displayed with pigment epithelium and photoreceptors to the top. Examples of sd-OCT

recordings before (left) and one week after (right) IAA (b, d, e) or PBS (a, c, f) application. a

PBS i.p. b IAA 65 mg/kg BW i.p.; red arrows indicate examples for the thickness

measurement in one retina. c PBS i.v. d IAA 60 mg/kg BW i.v. e IAA 2 x 30 mg/kg BW i.v.;

the red bars mark the outer nuclear layer, which is significantly thinner one week after

injection. f intravitreal injection of PBS. g, h Retinal thickness measurements based on sd-

OCT data 4 weeks after i.p. injection (g) and 2 weeks after i.v. injection (h) of IAA and PBS.

Values represent mean ± SD (for 2x30 mg/kg BW IAA: ****p<0.0001 vs. 0, 55, and 60

mg/kg BW, one-way ANOVA with Bonferroni’s post hoc test). i Retinal thickness

measurements under control conditions before and one week after i.p., i.v. and intravitreal

(IVI) injection of PBS.

Page 67: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

67

RESULTS

Figure 4 Confocal images of immunolabelled vertical sections obtained from retinae

prepared two weeks after systemic application. The white bars in a and c mark the

boundaries of the outer nuclear layer. Red: glutamine synthetase, Müller cells spanning nearly

the entire thickness of the retina; blue: PNA, cone outer segments and endfeet; green: GFAP.

a Control condition. b IAA i.v. 60 mg/kg BW. c IAA i.v 2x30 mg/kg BW (note that ONL

thickness and number of cones are reduced). d Glial reaction is indicated by GFAP expression

in Müller cell processes (same section as in C). e Rd10 mouse retina at the age of 50 days

displaying total loss of photoreceptors. OR: outer part of the retina; IR: inner part of the

retina; OS: outer segments; IS: inner segments; ONL: outer nuclear layer; OPL: outer

plexiform layer; INL: inner nuclear layer; IPL: inner plexiform layer; GCL: ganglion cell

layer.

Page 68: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

68

RESULTS

Figure 5 Effects of intravitreal injection of IAA. a Overview of effects. b Example of a

dense cataract seen one week after the injection of 0.5 mg/kg BW. c Phthitic eye two weeks

after injection of 0.5 mg/kg BW.

Figure 6 Retinal changes after intravitreal injection. OCT scans one and two weeks after

injection of 0.1 (a) and 0.15 mg/kg BW IAA (b). The scans were taken at the height of the

papilla (a) or at a position superior to the papilla (b). The scanning position for the cross

sectional image is indicated by the green line in the respective IR reflection image of the

fundus (b). Note that OCT scan appears blurry after two weeks making it difficult to

determine the retinal thickness accurately (position for thickness measurements indicated by

bars and double arrows).

Page 69: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

69

RESULTS

Figure 7 ERG after intravitreal IAA application. Examples of full-field ERGs recorded

before and one week after intravitreal (IVI) injection. a PBS. b 0.5 mg/kg IAA IVI. c, d

Changes in the ERG after IVI IAA application. Amplitude of a-waves (c) and b-waves (d)

corresponding to the concentration of IAA given.

Figure 8 Confocal images of immunohistochemically stained vertical sections through

the retina two weeks after injection of IAA into the vitreous. The borders of the ONL are

labelled by dashed lines. Stainings and abbreviations as in Fig. 4. a Untreated control. b 0.15

mg/kg BW IAA. c 0.2 mg/kg BW. d - f 0.25 mg/kg BW. Overall retinal thickness is reduced

in a variable way, but ONL thickness and cone density are only mildly affected. e Müller cells

express GFAP (same section as in d).

Page 70: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

70

RESULTS

5.3 STUDY III

SELECTIVE PHOTORECEPTOR DEGENERATION BY INTRAVITREAL INJECTION

OF N-METHYL-N-NITROSOUREA

Sarah Rösch1,2

, Sandra Johnen1, Anja Mataruga

3, Frank Müller

3, Christiane Pfarrer

2, and Peter

Walter1

1. Department of Ophthalmology, RWTH Aachen University, Germany

2. Department of Anatomy, University of Veterinary Medicine, Hannover Germany

3. Institute of Complex Systems, Cellular Biophysics, ICS-4, Forschungszentrum Jülich

GmbH, Germany

SUBMITTED/ UNDER REVIEW

Corresponding Author: Peter Walter, Department of Ophthalmology, RWTH Aachen

University, Pauwelsstr. 30, 52074 Aachen, Tel +49-241-8088191, fax +49-241-8082408, mail

to [email protected]

This manuscript is part of a doctoral thesis at the University of Veterinary Medicine,

Hannover, Germany (01.10.2013).

Word count: 4417

This work was supported by the DFG grants WA 1472/6-1 to PW and MU 3036/3-1 to FM.

Page 71: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

71

RESULTS

5.3.1 ABSTRACT

Purpose: To characterize the effects of intravitreal injections of N-Methyl-N-nitrosourea

(MNU) in comparison to its systemic application as a measure to induce unilateral

photoreceptor degeneration.

Methods: Eight weeks old male C57BL/6J mice received either intraperitoneal injections (3

animals) or intravitreal injections (24 animals) of MNU in different concentrations and were

observed in the following two weeks using electroretinography (ERG), spectral domain

optical coherence tomography (sd-OCT), and immunohistochemistry.

Results: The intraperitoneal application of MNU showed moderate systemic toxic effects,

indicated by a loss of body weight of 12% within the first two days. In both eyes the ERG

became extinguished and sd-OCT scans showed a thinning of the retina predominantly in the

outer nuclear layer (ONL). Immunohistochemistry demonstrated the selective loss of rods and

cones. Mice receiving intravitreal injections showed nearly no weight loss after the injections

and no reduction of their general welfare. After two weeks, in the injected eye, but not in the

control eye, ERG, sd-OCT, and immunohistochemistry revealed changes identical to those

seen after systemic application.

Conclusion: The intraperitoneal application of MNU led to moderate systemic side effects in

mice and to selective photoreceptor degeneration. Intravitreal injections of MNU also induced

photoreceptor degeneration, but without systemic side effects. This tool may be helpful in

larger species where genetic models of receptor degenerations are not applicable but where

the size of the eye is more suitable to study surgical or other approaches to treat blindness

causing receptor degeneration.

Page 72: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

72

RESULTS

5.3.2 INTRODUCTION

Inherited retinal dystrophies, such as Retinitis pigmentosa (RP), are important and frequent

causes for blindness and observed in approximately 1 of 4000 persons.1,2

Research efforts are

focused on the genetic background of these diseases, on the mechanisms of degeneration, but

also on possible treatment strategies. Genetic models in rodents expressing retinal

degeneration, e.g. the retinal degeneration (rd) 1 mouse, rd10 mouse3-13

, and the RCS rat

(Royal College of Surgeons)14-17

, are indispensable tools to explore experimental therapies for

currently untreatable conditions. In rd1 and rd10 mice, mutations in the gene encoding for the

ß-subunit of rod cGMP phosphodiesterase result in the described disease pattern with an in

contrast to human RP central to peripheral gradient.3-13

Because of the later onset and milder

retinal degeneration in contrast to the rd1 mouse, the rd10 mouse provides a better genetic

mouse model for human RP.12

In the RCS rat, a deletion in the gene encoding the receptor

tyrosine kinase (Mertk) expressed in retinal pigment epithelium (RPE) cells, leads to a

disturbed phagocytosis of photoreceptor outer segments and consequently to photoreceptor

degeneration by apoptosis.17

Regardless of the cause of outer retinal degeneration in the rd1

mouse, rd10 mouse, and RCS rat, a progressive loss of photoreceptors with age comparable to

RP is observed, which is due to apoptosis.18

The thickness of inner nuclear layers and of the

ganglion cell layer remains unaffected.12-17

However, cellular and organizational and

functional changes in the inner retina were also observed.7,19

Beside retinal transplants, stem

cell therapy, and gene therapy, retinal implants have been developed and are currently used in

humans. Further development and improvement of such implants require experimental

surgical approaches performed in larger animal species than in mice or rats, e.g., in pigs, cats

or rabbits exhibiting a specific loss of photoreceptors. Genetic models in larger animals are

difficult to utilize because of costs and duration of disease manifestation.20

Photoreceptor degeneration may also be induced by extrinsic physical measures such as light

exposure or ionizing radiation on the developing retina.22,23

Retinal degenerations are also

observed after systemically applied pharmaceuticals such as iodoacetic acid (IAA)19,24,25

or N-

methyl-N-nitrosourea (MNU).2,18,26-30

After systemic treatment MNU leads dose dependent to

extensive oxidative stress resulting in retinal photoreceptor cell death due to apoptosis, what

was confirmed by TUNEL staining.26-28

Photoreceptors can be rescued by several inhibitors

of apoptosis.27,28

The toxic effects of MNU on photoreceptors in a 7-day period after systemic

Page 73: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

73

RESULTS

application have been described in different animals.26-30

In mice and rats, a concentration of

60 mg/kg bodyweight (BW) MNU was effective when applied intraperitoneally, while in

rabbits the intravenous application of 40 mg/kg BW effectively caused photoreceptor

degeneration.26-30

Likewise to the genetic models in mice, the loss of photoreceptors

proceeded with a central to peripheral gradient, but instead of affection of first rods and

consecutively cones, after MNU treatment both rods and cones were lost simultaneously and

to the same extent.26-30

The number of cells within the inner nuclear layer and within the

ganglion cell layer did not decrease and the inner retinal neurons showed features of

morphological remodeling comparable to the genetic model.26-30

A side effect of systemic

MNU application is the induction of neoplasms.31-33

In rabbits after systemic intravenous

injection, 68.8% of the animals developed brain tumors.31

Furthermore, tumors frequently

occurred as small intestine carcinomas and as multiple vessel wall sarcomas in different

organs.31

In rats, mammary tumors or prostate tumors can be induced by a single

intraperitoneal or intravenous injection of 50 mg/kg BW MNU.32,33

Tumors grew 12-16

weeks after injection.31-33

We were motivated to identify a safe and reproducible procedure for inducing unilateral outer

retinal degeneration by intravitreal application of pharmaceuticals, avoiding systemic side

effects and keeping the contralateral eye as an intraindividual control eye. Therefore, we

investigated the effects of intravitreal injections of MNU on the mouse retina.

Page 74: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

74

RESULTS

5.3.3 MATERIALS AND METHODS

The materials and methods were performed as also described in Study I: ”Correlations

between ERG, OCT and anatomical findings in the rd10 mouse” and Study II: “The effects of

iodoacetic acid on the mouse retina”.

All experiments were performed in accordance with the ARVO declaration for the use of

animals in ophthalmic and vision research and in accordance with the German Law for the

Protection of Animals and after approval was obtained by the regulatory authorities. The

proposal of the animal experiments was approved by the LANUV (Landesamt für Natur,

Umwelt und Verbraucherschutz Nordrhein Westfalen, Recklinghausen, Germany; file

reference: 84-02.04.2011.A386).

Animals

Adult pigmented male wild type mice (C57BL/6J; 8 weeks) were housed in standard

conditions under 12/12-hour light-dark cycle with food and water available ad libitum.

Because of the possible environmental hazard due to the carcinogenic and teratogenic

potential of N-methyl-N-nitrosourea (MNU), the animals were kept in special one-way cages

(PET; Tecniplast Deutschland GmbH; Hohenpreißenberg; Germany) after each MNU

application under fume hoods with the same light and food conditions as before. The mice

received sterile N-methyl-N-nitrosourea (MNU; Sigma Aldrich, Germany) in different

concentrations intraperitoneally (3 animals, 60 mg/kg BW) or intravitreally (24 animals, 0.15

- 3 mg/kg BW). Because of its poor stability in aqueous solutions and the duration of the

procedure of intravitreal injection, MNU was dissolved in dimethylsulfoxide (DMSO; Serva,

Heidelberg, Germany) immediately before use and further diluted with sterile phosphate

buffered saline (PBS). Animals were examined five days before as well as 7 days and 14 days

after injection of MNU to monitor the effects of the injection on retinal morphology and

photoreceptor function. Body weight was recorded on a daily basis during the entire

experiment. Animals were sacrificed for histological examinations two weeks after treatment.

Mice were anesthetized with ketamine (70 mg/kg Ketamin® 10%, CEVA, Germany) and

xylazine (10 mg/kg Xylazin 2% Bernburg®, Medistar, Ascheberg, Germany) in 0.1 mL of

Page 75: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

75

RESULTS

saline for in vivo measurements, examinations and intravitreal injections. For the systemic

treatment with MNU no anesthesia was given.

Systemic MNU-application

MNU solution was administered intraperitoneally in three mice with a concentration of 60

mg/kg BW and pre-warmed at room temperature. This concentration was described to cause

photoreceptor degeneration in mice.23

Very rapid injections were avoided.

Intravitreal MNU-injection

Intravitreal injections were performed using a 27 gauge steel needle (BD Microlance TM

3,

Heidelberg, Germany) and a nanoliter injection system with a 3D micromanipulator

(Nanoliter 2000; World Precision Instruments, Inc. Sarasota, FL, USA). After puncture in the

dorso-nasal area of the limbus, 2 µL were injected with a glass capillary in an angle of 30°

(Fig. 8, yellow arrow demonstrates direction of injection). Preliminary control experiments

showed that the injection technique was safe and that the volume (defined volume of 2 µL)

could be placed in the vitreous cavity without touching the lens or even the retina. Different

concentrations of MNU were injected intravitreally diluted with DMSO and PBS. MNU was

injected intravitreally in the left eye of 24 mice (0.15, 0.2, 0.25, 0.3, 0.5, 1.0, 2.0, 3.0 mg/kg

BW; n=3 for each concentration). We used DMSO as first solvent to guarantee better

solubility of MNU and to minimize degradation of MNU during the time consuming

intravitreal injection.

All intravitreally injected mice were treated by antibiotic eye ointment (Polyspectran, Alcon

Pharma GmbH, Freiburg, Germany) to avoid inflammatory reactions up to three days after

injection.

Control Interventions

Two wild type mice were kept under the same husbandry condition to evaluate systemic

effects of changing the cage conditions from normal cages to the one-way system. Further

control animals received intraperitoneal sham injections (PBS (n = 3) or DMSO + PBS

(n = 3)).

Page 76: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

76

RESULTS

Intravitreal control injections of PBS (2 µL), DMSO (maximal volume of 1 µL diluted to 2

µL with PBS) and fluorescein (different concentrations dissolved in 2 µL of PBS) were

performed to confirm the safety of the injection technique. Absence of leakage from the

injection area directly after injection of fluorescein was confirmed. Therefore, images were

taken by a confocal laser scanning microscope (Heidelberg Retina Angiograph-1, Heidelberg

Engineering, Heidelberg, Germany) in the fluorescein angiography mode.

All control animals (PBS; DMSO + PBS) were examined using the same methods and time

schedule as the experimental animals.

Electroretinogram recordings

After a dark adaption of one hour, pupils were dilated with one drop of Tropicamide 2.5% eye

drops (Pharmacy of the University Hospital Aachen, Germany). Mice were placed in front of

a full-field dome stimulator. The eyes were locally anesthetized with

proxymetacainhydrochloride 0.5% eye drops (Proparakain- POS®, Ursapharm, Saarbrücken,

Germany), and a goldring electrode (animal electrode 0.5mm ø 3mm Roland Consult,

Brandenburg an der Havel, Germany) was placed on the corneal surface of each eye. Non-

irritating artificial tears were used serving for corneal clarity and for a better ionic conduction

(Methylcellulose, Methocel® 2%, Omni Vision, Puchheim, Germany). A goldring electrode

in the mouth (contact with the mouth mucosa) served as reference electrode and a

subcutaneous silver needle electrode in the lumbar region as ground electrode.

Electroretinography was performed using the Reti System designed for rodents (Roland

Consult Electrophysiological Diagnostic Systems, Brandenburg, Germany). Mice were

examined by full-field ERGs according to the ISCEV standard protocol with five light stimuli

per recording.35

Amplitudes of the major ERG components, e.g., a- and b-wave, and implicit

times of rod and cone responses per recording were determined by averaging the responses of

the five light stimuli.

Spectral domain Optical Coherence Tomography

After anesthesia mice were positioned in front of the Spectralis® OCT system (Heidelberg

Engineering, Heidelberg, Germany) to perform the spectral domain Optical Coherence

Tomography. The scanning system was prepared additionally by a +25 D lens to correct for

Page 77: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

77

RESULTS

rodent optics. The optic disc was located as main landmark and retinal thickness

measurements were conducted using the cross sectional image centered on the optic disc. The

total retinal thickness as well as the thickness of the different layers was measured at six

positions across the retina (Fig. 3B, red arrows). Thickness values of each eye were averaged.

For documentation purposes, confocal images of the fundus of the evaluated eyes were

recorded (IR reflection image, wavelength 715nm). To further document possible

macroscopic changes of the treated or non-injected contralateral eyes, pictures of the mice

were taken by a Nikon D80 camera (Nikon AF Micro Nikkor 105 mm, 2896 Pixel x 1944

Pixel x 24 Bit).

Histology

After a duration of in vivo experiments of two weeks, mice were euthanized by isoflurane

(Forene 100% (V/V)®, Abott GmBH, Wiesbaden, Germany) overdosing and decapitated.

Eyes were processed in immunohistochemistry as well as in Hematoxylin and Eosin stainings.

Immunohistochemistry was performed as described earlier by Mataruga et al. (2007).37

In

brief, eyes were enucleated and opened by an encircling cut at the limbus. The retinae in the

eyecup were immersion-fixed for 30 minutes in 4% paraformaldehyde (PA) in 0.1 M

phosphate buffer (PB) at room temperature and washed in PB several times. Tissue was

incubated in 10% sucrose/PB for 1 hour, followed by 30% sucrose overnight. The retina was

flat embedded and frozen in optimal cutting temperature (OCT) compound (NEG-50, Richard

Allen Scientific, Thermo Fisher Scientific, Germany). Vertical sections (20 μm thickness)

were cut on a cryostat (HM 560 CryoStar; MICROM; Walldorf; Germany) and collected on

Superfrost Plus slides (Menzel, Braunschweig, Germany). Sections were pre-treated with

blocking solution (5% chemiblocker (Chemicon, Hofheim, Germany), 0.5% Triton-X100 in

PB, and 0.05% NaN3) for 1 hour, followed by incubation with primary antibodies over night,

diluted in the same solution. Following primary antibodies were used: anti-GFAP (anti glial

fibrillary acidic protein; raised in chicken, 1:2000; Novus, Germany), antibodies against

glutamine synthetase (raised in mouse, 1:4000; BD Biosciences, Germany), anti-CabP (anti

calbindin 28K; raised in mouse, 1:1000; Sigma, Germany), anti-PKC (anti protein kinase C α;

raised in rabbit, 1:4000; Santa Cruz, Heidelberg Germany), against Calretinin (AB1550,

raised in goat, 1:3000; Millipore, Germany), against HCN1 (RTQ-7C3, raised in rat, 1:10; F.

Page 78: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

78

RESULTS

Müller, Forschungszentrum Jülich, Germany), against recoverin (AB5585, raised in rabbit,

1:2000; Millipore, Germany), and against rhodopsin (1D4, 1:500; R.S. Molday, British

Columbia, Canada). Sections were washed in PB and incubated in secondary antibodies

diluted in 5% Chemiblocker, 0.5% Triton-X100 in PB for 1 hour, washed in PB and

coverslipped with Aqua Polymount (Polysciences, Eppelheim, Germany). Secondary

antibodies included: donkey anti-chicken Cy2 (1:400; Dianova, Germany), donkey anti-

mouse Cy3 (1:100; Dianova, Germany), donkey anti-rabbit Cy2 (1:400; Dianova, Germany),

donkey anti-goat Alexa647 (1:200; Invitrogen, Germany), donkey anti-rat Cy3 (1:500;

Dianova, Germany), and donkey anti-mouse Dy649 (1:500; Dianova, Germany). The lectin

Peanut agglutinin (PNA, biotinylated, 1:1600; Sigma Aldrich; Germany) was visualized using

StreptavidinAlexa647 (1:100; Invitrogen, Germany).

Sections were examined with a Leica TCS confocal laser scanning microscope (Leica

Microsystems, Heidelberg, Germany) with 63x/1.4 oil immersion lenses. Images were

processed and printed with Adobe Photoshop. For triple labeling, primary antibodies were

mixed and applied simultaneously. All secondary antibodies were highly cross-absorbed and

were carefully tested to exclude reactions with the wrong primary antibody. Concentration of

the antibodies, laser intensity, and filter settings were carefully controlled and the sequential

scanning mode was employed to completely rule out cross-talk between the fluorescence

detection channels. Band pass filters of 500 - 530 nm for green fluorescence (Cy2), 580 - 650

nm for red fluorescence (Cy3), and 680 - 750 nm for infrared fluorescence (Alexa647) were

used.

For Hematoxylin-Eosin staining, eyes were enucleated, punctured three times at the limbus

and fixed by immersion as described above. Eyes were dehydrated in a tissue dehydration

automat (mtm, Slee, Mainz, Germany) by incubation in a series of increasing ethanol

concentrations (2 times x 70%, 2 x 96%, 3 x 100% for one hour), followed by xylene (3 times

1 hour) and paraffin (4 times one hour), and embedded in paraffin. Sections of 5 μm thickness

were cut with a microtome (R. Jung, Heidelberg, Germany), collected on slides,

deparaffinized, rehydrated, and stained with Hematoxylin and Eosin. Pictures were taken

using a Leica DMRX microscope (Leica Microsystems, Heidelberg, Germany).

Page 79: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

79

RESULTS

The thickness of the entire retina and the individual retinal layers was determined in both

immunohistochemical and in Hematoxylin-Eosin stainings at six positions close to the papilla.

For each retina, thickness values obtained from these positions were averaged.

Urine test

Before, two and three hours, one and two days after systemic and intravitreal application of

MNU urine tests (Ultra performance liquid chromatography (UPLC); Aquity; Waters,

Eschborn, Germany) were made to test the excretion of MNU for safety reasons.

Page 80: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

80

RESULTS

5.3.4 RESULTS

Systemic application of MNU. Weight Loss. Intraperitoneal application of MNU resulted in

systemic side effects in mice with a weight loss of about 12% within the first two days. In

Figure 1 the weight loss after different forms of treatment with MNU is demonstrated to

reflect the well-being under changed husbandry conditions in one way cages.

Electroretinography. Systemically administered MNU led to an extinction of the scotopic

(Figure 2C - E) and photopic (Figure 3C) ERG responses after one and two weeks.

Optical Coherence Tomography. One week after intraperitoneal MNU application, a

reduction in total retinal thickness was observed, while the space between the retina and the

RPE seemed to be filled with cell debris (Figure 4C). After two weeks the debris had

disappeared completely, while the reduction of retinal thickness was more pronounced as

demonstrated in Figure 4C. Two weeks after injection, the inner nuclear layer became

adjacent to the pigment epithelium (Figure 4C).

Microscopy. Histological analysis confirmed the reduction of retinal thickness as seen in sd-

OCT recordings in-vivo with the pronounced loss of photoreceptors and the thinning of the

outer nuclear layer (ONL) after MNU treatment. Müller cells labeled with anti-GFAP (Figure

5D) had become reactive as seen also during other causes of retinal degeneration.36

Under

normal conditions, GFAP is only expressed in astrocytes. Representative sections are shown

in Figure 5. In animals that had received vehicle injection only (Figure 5B), the retina

remained unaffected. In the animals treated intraperitoneally with MNU, the ONL was

reduced to no or only one row of photoreceptor somata (Figure 5C), while the thickness of the

inner retina was statistically not different from the thickness in control eyes (Figure 8C). The

thinning of the outer retina was reminiscent of the photoreceptor loss observed in Retinitis

pigmentosa (RP) or in late stages of rodent models of RP such as in the rd10 mouse (Figure

5E).6-14

Intravitreal injection of MNU. Weight loss. Animals that received PBS, DMSO + PBS or

MNU as intravitreal injections showed no remarkable weight loss or other systemic side

effects (see Figure 1).

Electroretinography. The control interventions – injection of PBS or DMSO + PBS – as well

as the intravitreal injection of 0.15 up to 0.5 mg/kg BW MNU had no effect on the ERG. In

Page 81: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

81

RESULTS

animals treated with 1 and 2 mg/kg BW, scotopic (Figure 6) and photopic (Figure 3) ERGs

were affected. For both concentrations, in two of three animals the amplitudes of the a-wave

and the b-wave in the scotopic ERG were diminished and in one animal the ERG was

completely abolished. In all animals injected intravitreally with 3 mg/kg BW the scotopic

(Figure 6) and photopic (Figure 3) ERG was completely extinguished.

Optical Coherence Tomography. Sd-OCT measurements one week after the injection of 1, 2,

and 3 mg/kg BW revealed cell debris between the inner retina and the pigment epithelium,

similar to the findings observed after intraperitoneal application (compare Figure 7D - F to

Figure 4C). In animals that had received 3 mg/kg BW MNU, the somata of the photoreceptors

(outer nuclear layer, ONL) had disappeared throughout the entire retina. Upon injection of 1

or 2 mg/kg BW MNU, the cell death in the ONL was not homogeneous. In the superior

peripheral retina, i.e. close to the injection site, most or all of the cell layers in the ONL had

vanished, while in the inferior retina the reduction of ONL thickness was less than 50%.

Microscopy. The reduction of retinal thickness upon intravitreal injection of 1, 2, and 3 mg/kg

BW MNU observed in the OCT was verified by histology (Figure 8). MNU treatment led to a

thinning of the ONL, while the thickness of the inner retina was not significantly affected

(Figure 8). In animals that had received 1 or 2 mg/kg BW MNU, immunohistochemical data

(Figure 8, 9) confirmed that the degree of ONL thinning depended on the distance to the

injection site. In contrast to these findings, intravitreal injection of 3 mg/kg BW MNU

resulted in a complete loss of the ONL throughout the entire retina. All images shown in

Figure 8 are taken close to the papilla.

In Figure 9 superior parts (close to injection side) and inferior parts of the retina are illustrated

(Figure 9B, D, F and G) with a corresponding staining for rods and cones in each area (C, E,

G, I). The retinal areas with the more pronounced thickness loss in the ONL after treatment

with 1 and 2 mg/kg BW MNU (9B, D), are more close to the injection side (superior retina),

what is documented by the staining of red/green opsin (green). Red/green opsin is located in

the superior retina of mice. In the areas, where more layers of the photoreceptor somata

(ONL) remained, blue opsin staining (red) is represented, indicating an inferior retinal area.

The stainings in Figure 9 allowed the evaluation of the morphology of the photoreceptors

which remained after application of 1 or 2 mg/kg BW MNU. In surviving rods and cones, we

could observe following signs of degeneration (Figure 9). In Figure 9 B, F, D and H

Page 82: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

82

RESULTS

antibodies against rhodopsin and recoverin were used. Rhodopsin is presented in rod outer

segments is visualized in blue, while recoverin that is found throughout the entire

photoreceptors is shown in green. Rod outer segments, therefore, appear cyan. They are

shorter than normal in all sections shown, but are longest in regions most remote from the

injection site (Figure 9F, 1 mg/kg BW MNU, inferior part of the retina).

Cones also show features of degeneration. In Figure 9G (1 mg/kg BW MNU, inferior retina)

cones survived best, showing long outer segments stained in red for blue opsin. Closer to the

injection site (Figure 9C, 1 mg/kg BW MNU, superior retina) two abnormal features can be

observed. First, red/green opsin staining (green) is found throughout all cone compartments,

while normally only small amounts of opsin can be observed outside the outer segment.

Second, the outer segments are short and round. Close to the injection side after 2 mg/kg BW

MNU (Figure 9E), cones are completely gone (green staining represents background in inner

retinal cells), and in the referring inferior part of the retina (Figure 9I), outer segments (red)

are extremely shortened. Additionally to the photopic ERGs in Figure 3, in the same figure

immunohistochemically stained sections of intravitreally injected animals are illustrated to

underline the simultaneous death of rods and cones. In the photopic ERG cone responses are

comparably lost as well as rod driven responses (Figure 3).

In animals treated with 1 and 2 mg/kg BW MNU the stainings of HCN1 channels (red) and

recoverin in type 2 bipolar cells (green) do not reveal changes in the inner retinal layers

(Figure 9B, D, F, and H). To determine in detail the effects of MNU on inner retinal cells

additional stainings were performed. The first column of Figure 10 (A, F, K) shows

immunohistochemical stainings in wild type retina: Calbindin 28K (Figure 10A) is strongly

expressed in horizontal cell somata and their dendrites (upper cells, arrow) and weakly in

certain amacrine cells (lower part of the section). Rod bipolar cells (Figure 10F) can be

stained with antibodies against PKCα. Rod bipolar somata are usually elongated, axons are

clearly visible and axon terminals are compact. Certain amacrine cell types and three typical

bands formed by their dendrites in the IPL can be stained with antibodies against calretinin

(Figure 10K).

Upon intraperitoneal application of MNU (Figure 10B, G, L), fine horizontal cell dendrites

were lost as the presynaptic photoreceptors had disappeared, but horizontal cell somata were

mostly unchanged (Figure 10B). Some rod bipolar somata (Figure 10G, arrow) were found

Page 83: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

83

RESULTS

that were more roundish than usual, but most rod bipolar cells were unaffected. Amacrine

cells appeared unchanged (Figure 10L).

Upon intravitreal injection of 1 mg/kg BW (Figure 10C, H, M), 2 mg/kg BW (Figure 10D, I,

N) or 3 mg/kg BW MNU (Figure 10E, J, O), the following results were obtained. In those

regions of the retina where photoreceptors survived (1 and 2 mg/kg BW MNU), horizontal

cells (Figure 10C, D), rod bipolar cells (Figure 10H, I), and amacrine cells (Figure 10M, N)

were basically unaffected. The situation was different in those retinal regions where all

photoreceptors had disappeared, i.e. throughout the entire retina upon injection of 3 mg/kg

BW MNU (Figure 10E, J, O) and close to the injection site upon injection of 1 or 2 mg/kg

BW MNU (not shown). Here, horizontal cells were missing (Figure 10E), rod bipolar cell

somata became roundish (Figure 10J, arrow), their axons and axon terminal systems were

disorganized and more amacrine cells than usual expressed PKC (Figure 10J, arrowhead). The

calretinin positive bands in the IPL were slightly broader (Figure 10O).

In no case, the intravitreal injection affected the non-injected eye of the animal.

In none of the urine test samples MNU-molecules were found (data not shown).

Page 84: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

84

RESULTS

5.3.5 DISCUSSION

Genetic animal models of retinal degenerations and dystrophies, such as Retinitis pigmentosa

(RP) revealing selective photoreceptor degeneration, are important to study the underlying

disease mechanisms and also to establish possible treatment approaches. Several rodent

models are well established.3-17

In larger animals which would be more suitable to work on

surgical methods, genetic models are intrinsically expensive and disease progression is

slow.20

However, photoreceptor degeneration may also be induced pharmacologically and

may have the advantage that it can be induced in any wanted animal species.24-30

As one

possible pharmacological agent N-methyl-N-nitrosourea (MNU) was reported to lead to a

blindness causing selective photoreceptor cell apoptosis via oxidative stress when

administered intraperitoneally or intravenously.26-30

The resulting histological picture is well

analyzed in detail by different researchers26-30

and very similar to that presented in human

RP.26

However, retinal degeneration induced by systemic application of MNU not only leads

to bilateral disease, but also to a reduction of the general health status of the experimental

animals. Beside short-term effects caused by the toxicity of the substance, the induction of

tumors was described as long-term effect in rabbits and rats after systemic treatment with

MNU due to its DNA alkylating mode of action.31-33

Therefore, we proved to induce

photoreceptor degeneration in only one eye of mice by local administration of MNU and

avoiding systemic side effects.

Puthussery and Fletcher36

and Notomi et al.37

demonstrated, that intravitreally injected

adenosine triphosphate (ATP) induces photoreceptor degeneration by apoptosis, too.

However, ATP also leads to TUNEL-positive apoptotic events in the ganglion cell layer after

intravitreal application. 36

ATP was described to kill ganglion cells by excessive calcium

influx by stimulation of the P2RX7 receptor.37

In contrast to the effects of ATP on ganglion

cells, Tsuruma et al. reported, that MNU leads to radical production only in retinal cell

cultures of murine 661W photoreceptor-derived cells, but not in RGC-5, a mouse ganglion

cell line, and hence induces cell death specifically in retinal photoreceptor cells.28

The reason

why MNU toxicity is specific for photoreceptors and maybe also for horizontal cells, remains

unclear. MNU given intravitreally needs to penetrate through the retina or needs to be

transported through it to affect the outer retina. It is likely that photoreceptors are particularly

Page 85: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

85

RESULTS

susceptible due to its extreme high metabolic turnover in the photoreceptor outer segment /

RPE unit. Further experiments are needed to clarify the mode of MNU action.

Beside the intravitreal injections of MNU, the systemic application given as an intraperitoneal

injection served as our positive control. In these animals the well-being of the animals was

reduced for some hours after administration. They showed a reduction of weight with the

highest loss two days after injection. However, all animals survived. Regarding the outcome

of systemic MNU application, a rapid and nearly complete denudation of the ONL was

observed. Cells of the inner retina like horizontal, bipolar or amacrine cells were only slightly

affected. As the presynaptic photoreceptors were lost, horizontal cells and rod bipolar cells

lost most of their dendrites similar to findings described earlier.30

Because of the small vitreous space of the mouse eye and the proportionally large lens,

intravitreal injections in mice are difficult. To optimize the injection technique, we used a 3D

micromanipulator and nanoliter injection system. Injections of PBS, fluorescein, and

DMSO+PBS mixture, were used as negative controls, and could point out that the injection

procedure was safe. Examination of control animals succeeded without any changes in

functionality in vivo and showed no histological abnormalities. In contrast to the systemic

application, the well-being of the animals was only minimally affected upon intravitreal

injections of MNU, comparable to the welfare after an anesthetic intervention.

One week after intravitreal injection of 1, 2 or 3 mg/kg BW MNU, the OCT showed similar

changes as observed one week after intraperitoneal application. However, two weeks after 1

and 2 mg/kg BW MNU, not all somata of photoreceptors in the outer nuclear layer had

disappeared completely. The degree of photoreceptor degeneration depended on the distance

to the injection site. While at the point of injection in the superior retina no row of somata in

the ONL was left, five to ten rows of photoreceptor somata were detectable in the inferior

retina (Figure 9). Detailed analysis using photoreceptor markers (Figure 9) indicated that the

surviving photoreceptor population comprised both rods and cones, but that outer segments

were shorter than usual. Standard full-field ERG waves in animals injected with 1 and 2

mg/kg BW MNU under scotopic and photopic conditions were either diminished (2 animals

in each group) or even extinguished (one animal in each group, n=3; Figure 5D, E).

Upon injection of 3 mg/kg BW MNU, the photoreceptors (ONL) had disappeared throughout

the entire eye, while the inner retinal layers remained. Inner retinal layers of intravitreally

Page 86: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

86

RESULTS

injected animals (1, 2, or 3 mg/kg BW) showed no significant thickness differences to

uninjected eyes. The histological picture was reminiscent of the results obtained after

systemic treatment with MNU and results obtained with the genetic model (rd10) at postnatal

day 50 (Figure 8). Immunohistochemistry performed two weeks after injection revealed no

surviving photoreceptor population. In agreement with this finding, no responses could be

observed in the scotopic and photopic ERG two weeks after injection of MNU.

However, in those regions of intravitreally injected eyes where photoreceptors had vanished

entirely, also the horizontal and rod bipolar cells seemed to be affected. Two weeks after

injection, we noticed a complete loss of horizontal cell somata and processes in the retinal

regions where all photoreceptors had disappeared. While after systemic MNU treatment a loss

of horizontal cell processes in a study over three month has been described,30

a complete cell

loss has not been observed. In comparison, in rd10 mouse at the age of 9 months around 29%

of horizontal cell somata were lost.7

The rod bipolar cells seemed to be disorganized after an intravitreal injection of 3 mg/kg BW

MNU. For rd10 mice up to 45 days of age, no changes in the morphology, size, and

complexity of the axonal terminal systems of rod bipolar cells were found, but between 1.5

and 3.5 months of age the number or rod bipolar cells decreased by 20%.7

There are two ways how the effect of MNU on horizontal and rod bipolar cells can be

explained. First, at higher concentrations of MNU as they are reached at the site of injections

when using 1 or 2 mg/kg BW MNU or throughout the eye when using 3 mg/kg BW, MNU

might become toxic to horizontal cells and rod bipolar cells. Tsuruma et al. reported about a

decrease in thickness of the INL after intraperitoneal treatment with 75 mg/kg BW instead of

60 mg/kg BW, what may be based on a destruction of horizontal and rod bipolar cells, too.28

Second, the rapid loss of all photoreceptors at a retinal location within two weeks might have

a more pronounced effect on the survival of postsynaptic cells than the degeneration

occurring at a slower pace in the genetic model. After intravitreal injection of ATP also

degeneration of photoreceptors in variable degree is described, ranging from the destruction

of photoreceptor segments to a complete loss of retinal pigment epithelium (RPE) and in

regions of rapid and strong destruction also to highly disturbed synapses of the outer

plexiform layer.36

Page 87: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

87

RESULTS

In Summary, our results clearly indicate that MNU given intravitreally is a per se working

model for the induction of photoreceptor degeneration to simulate disease processes as seen in

RP or similar conditions. The mouse model has the shortcomings of a small eye. It was

difficult to achieve a homogeneous degeneration of photoreceptors throughout the retina upon

one intravitreal application. Photoreceptor loss was more pronounced close to the injection

site. This problem may not be encountered in larger eyes, such as in the rabbit, as injections of

MNU (1 - 2 mg/kg BW) at multiple positions in the large vitreal volume might result in a

more homogeneous elimination of photoreceptors. Based on these results, we would suggest

the use of MNU injections into the vitreous of mice or larger animals as a pharmaceutical

model to specifically induce degeneration of photoreceptors while maintaining an

intraindividual control eye.

5.3.6 ACKNOWLEDGEMENT

The authors thank Prof. Dr. R. Tolba and the institute of laboratory animal science of the

University Hospital RWTH Aachen for the great opportunity, for the access to one of their

laboratories, and the possibility to perform our experiments under special safety guards. We

further thank Christoph Aretzweiler for the excellent support in preparation and staining of

the eyes.

Page 88: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

88

RESULTS

5.3.7 REFERENCES

1. Berson EL. Retinitis pigmentosa. The Friedenwald Lecture. Invest Ophthalmol Vis Sci.

1993;34:1659-1676.

2. Tsubura A, Yoshizawa K, Kiuchi K, Moriguchi K. N-Methyl-N-nitrosourea-induced

Retinal Degeneration in Animals. Acta Histochem Cytochem. 2003;36:263-270.

3. Delyfer MN, Forster V, Neveux N, Picaud S, Léveillard T, Sahel JA. Evidence for

glutamate-mediated excitotoxic mechanisms during photoreceptor degeneration in the

rd1 mouse retina. Mol Vis. 2005;11:688-696.

4. Hart AW, McKie L, Morgan JE, et al. Genotype-phenotype correlation of mouse pde6b

mutations. Invest Ophthalmol Vis Sci. 2005;46:3443-3450.

5. Ahuja S, Ahuja-Jensen P, Johnson LE, et al. Rd1 Mouse retina shows an imbalance in the

activity of cysteine protease cathepsins and their endogenous inhibitor cystatin C.

Invest Ophthalmol Vis Sci. 2008;49:1089-1096.

6. Gibson R, Fletcher EL, Vingrys AJ, Zhu Y, Vessey KA, Kalloniatis M. Functional and

neurochemical development in the normal and degenerating mouse retina. J Comp

Neurol. 2013;521:1251-1267.

7. Gargini C, Terzibasi E, Mazzoni F, Strettoi E. Retinal organization in the retinal

degeneration 10 (rd10) mutant mouse: a morphological and ERG study. J Comp

Neurol. 2007;500:222-238.

8. Chen M, Wang K, Lin B. Development and degeneration of cone bipolar cells are

independent of cone photoreceptors in a mouse model of retinitis pigmentosa. PLoS

ONE 2012;7:e44036.

9. Samardzija M, Wariwoda H, Imsand C, et al. Activation of survival pathways in the

degenerating retina of rd10 mice. Exp Eye Res. 2012;99:17-26.

10. Chrenek MA, Dalal N, Gardner C, et al. Analysis of the RPE sheet in the rd10 retinal

degeneration model. Adv Exp Med Biol. 2012;723:641-647.

11. Cang B, Hawes NL, Hurd RE, Davisson MT, Nusinowitz S, Heckenlively JR. Retinal

degeneration mutants in the mouse. Vision Res. 2002;42:517-525.

12. Chang B, Hawes NL, Pardue MT, et al. Two mouse retinal degenerations caused by

missense mutations in the beta-subunit of rod cGMP phosphodiesterase gene. Vision

Res. 2007;47:624-633.

Page 89: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

89

RESULTS

13. Pang JJ, Boye SL, Kumar A, et al. AAV-mediated gene therapy for retinal degeneration in

the rd10 mouse containing a recessive PDEbeta mutation. Invest Ophthalmol Vis Sci.

2008;49:4278-4283.

14. Hess HH, Newsome DA, Knapka JJ, Westney GE. Slitlamp assessment of age of onset

and incidence of cataracts in pink-eyed, tan-hooded retinal dystrophic rats. Curr Eye

Res. 1982;2:265-269.

15. Matuk Y. Retinitis pigmentosa and retinal degeneration in animals: a review. Can J

Biochem Cell Biol. 1984;62:535-546.

16. Fletcher EL, Kalloniatis M. Neurochemical development of the degenerating rat retina. J

Comp Neurol. 1997;388:1-22.

17. Perche O, Doly M, Ranchon-Cole I. Transient protective effect of caspase inhibitors in

RCS rat. Exp Eye Res 2008;86:519-527.

18. Nambu H, Yuge K, Nakajima M, et al. Morphologic characteristics of N-mehtyl-N-

nitrosourea-induced retinal degeneration in C57BL mice. Pathol Int. 1997;47:377-383.

19. Liang L, Katagiri Y, Franco LM, et al. Long-term cellular and regional specificity of the

photoreceptor toxin, iodoacetic acid (IAA), in the rabbit retina. Vis Neurosci.

2008;25:167-177.

20. Tso MO, Li WW, Zhang C, Lam TT, Hao Y, Petters RM, Wong F. A pathologic study of

degeneration of the rod and cone population of the rhodopsin Pro347Leu transgenic

pigs. Trans Am Ophthalmol Soc 1997;95:467-479.

21. Jae S, Ahn K, Kim J, Seo J, Kim H, Goo Y. Electrophysiological and histologic

evaluation of the time course of retinal degeneration in the rd10 mouse model of

retinitis pigmentosa. Korean J Physiol Pharmacol. 2013;17:229-235.

22. Harada T, Harada C, Sekiguchi M, Wada K. Light-induced retinal degeneration

suppresses developmental progression of flip-to-flop alternative splicing in GluR1. J

Neurosci. 1998;18:3336-3343.

23. Shively JN, Phemister RD, Epling GP, Jensen R. Pathogenesis of radiation-induced retinal

dysplasia. Invest Ophthalmol. 1970;9:888-900.

24. Scott P, Kaplan H, Sandell J: Anatomical evidence of photoreceptor degeneration induced

by iodoacetic acid in the porcine eye. Exp Eye Res. 2011;93:513-527

25. Noell WK, Experimentally induced toxic effects on structure and function of visual cells

and pigment epithelium. Am J Ophthalmol. 1953;36:103-116.

Page 90: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

90

RESULTS

26. Tsubura A, Yoshizawa K, Kuwata M, Uehara N. Animal models for retinitis pigmentosa

induced by MNU; disease progression, mechanisms and therapeutic trials. Histol

Histopathol. 2010;25:933-944.

27. Yoshizawa K, Yang J, Senzaki H, et al.Caspase-3 inhibitor rescues N-methyl-N-

nitrosourea-induced retinal degeneration in Sprague-Dawley rats. Exp Eye Res.

2000;71:629-635.

28. Tsuruma K, Yamauchi M, Inokuchi Y, Sugitani S, Shimazawa M, Hara H. Role of

oxidative stress in retinal photoreceptor cell death in N-mehtyl-N-nitrosourea-treated

mice. J Pharmacol Sci. 2012;118:351-362.

29. Herrold KM. Pigmentary degeneration of the retina induced by N-methyl-N-nitrosourea.

An experimental study in syrian hamsters. Arch Ophthalmol. 1967;78:650-653.

30. Nagar S, Krishnamoorthy V, Cherukuri P, Jain V, Dhingra NK. Early remodeling in an

inducible animal model of retinal degeneration. Neuroscience 2009;160:517-529.

31. Schreiber D, Jänisch W, Warzok R, Tausch H. Induction of brain and spinal cord tumors

in rabbits with N-methyl-N-nitrosourea. Z Gesamte Exp Med. 1969;150:76-86.

32. Alfaro Y, Delgado G, Cárabez A, Anguiano B, Aceves C. Iodine and doxorubicin, a good

combination for mammary cancer treatment: antineoplastic adjuvancy,

chemoresistance inhibition, and cardioprotection. Mol Cancer. 2013 doi:

10.1186/1476-4598-12-45.

33. McCormick DL, Rao KV, Dooley L, et al. Influence of N-methyl-N-nitrosourea,

testosterone and N-(4-hydroxyphenyl)-all-trans-retinamide on prostate cancer induction

in Wistar-Unilever Rats. Cancer Res.1998;58:3282-3288.

34. Marmor MF, Fulton AB, Holder GE, Miyake Y, Brigell M, Bach M. ISCEV Standard for

full-field clinical electroretinography (2008 update). Doc Ophthalmol. 2009;118:69-77.

35. Eisenfeld AJ, Bund-Milam AH, Sarthy PV. Müller cell expression of glial fibrillary acidic

protein after genetic and experimental photoreceptor degeneration in the rat retina.

Invest Ophthalmol Vis Sci. 1984;25:1321-1328.

36. Puthussery T, Fletcher E. Extracellular ATP Induces Retinal Photoreceptor Apoptosis

through Activation of Purinoceptors in Rodents. J Comp Neurol. 2009;513:430-440.

37. Notomi S, Hisatomi T, Kanemaru T, Takeda A, Ikeda Y, Enaida H, Kroemer G, Ishibashi

T. Critical involvement of extracellular ATP acting on P2RX7 purinergic receptors in

photoreceptor cell death. Am J Pathol 2011;179:2798-2809.

Page 91: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

91

RESULTS

5.3.8 FIGURES

Figure 1. Side effects of MNU application. Loss of body weight as an indicator of general

well-being after application of MNU given intraperitoneally (i.p.) and intravitreally (IVI) in

comparison to untreated animals and control animals in the same changed husbandry

conditions. ***p<0.003 vs. i.p. MNU application, one-way ANOVA with Bonferroni’s post

hoc test. (MNU i.p.: n=3; Control DMSO/PBS i.p.: n=3; untreated: n=2; intravitreal (IVI)

each concentration: n=3).

Page 92: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

92

RESULTS

Figure 2. Scotopic ERG after systemic MNU application. (A) - (C) Examples of scotopic

full-field ERG recordings from mice before (black line) and one week after (grey line)

systemic intraperitoneal application of PBS (A), DMSO + PBS (B), and 60 mg/kg BW MNU

(C) under the same husbandry conditions. Scotopic 3 cds/m2, 0.067 Hz. In (C) an extinction

of the ERG curve one week after application could be observed. (D), (E) Amplitudes of a-

wave (D) and b-wave (E) before and two weeks after i.p. MNU application (n=3) at a light

intensity of 3 cds/m2 in the scotopic ERG. Mean + SD. DMSO + PBS (n=3) and PBS (n=3)

injections served as controls (both eyes per n).

Page 93: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

93

RESULTS

Figure 3. Photopic ERG after MNU application and confocal images of

immunohistochemically stained vertical retinal sections. The a-wave and b-wave of

animals in the photopic ERG (3 cds/m2; 0,625 Hz) was diminished one week after

intraperitoneal (60 mg/kg BW, (B)) and intravitreal (C) - (E) treatment with MNU. The

contralateral eye (CL3) of the animal treated with 3 mg/kg BW revealed a normal photopic

ERG (E). The immunohistochemically stained sections were used to demonstrate the

simultaneous degeneration of rods and cones after intraperitoneal and intravitreal injection of

MNU in different concentrations (F). Stainings against red/green opsin (green, cone outer

segments in the superior retina), blue opsin (red, cone outer segments in the inferior retina)

and PEA (cones, blue) were used. The involvement of cones in the degenerative process is

seen in the photopic ERG and in histology by missing outer segments and an unusual

morphology.

Page 94: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

94

RESULTS

Figure 4. Thickness evaluation in the OCT. Retinal images and thickness obtained by sd-

OCT measurements. Left panel. All images are taken in the height of the optic disk and

displayed with pigment epithelium and photoreceptors to the top. Examples of sd-OCT

recordings before (left), one week after (middle), and two weeks after (right) intraperitoneal

PBS (A), DMSO + PBS (B), or MNU (C) application. Red arrows (B) indicate examples for

the thickness measurement in one retina. (C) MNU 60 mg/kg BW i.p.; the red bars mark the

outer nuclear layer (ONL), which is significantly thinner one and two weeks after injection;

enlargement of the area next to the optic disk shows the destruction of the ONL and the debris

in the area of the ONL. (D) Retinal thickness measurements based on sd-OCT data before,

one week, and two weeks after i.p. injection of PBS, DMSO + PBS and MNU (n=3). Values

represent mean ± SD (for 60 mg/kg BW one and two weeks after application: ****p<0.0001

vs. the controls, one-way ANOVA with Bonferroni’s post hoc test).

Page 95: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

95

RESULTS

Figure 5. Confocal images of immunolabelled vertical sections obtained from retinae

prepared two weeks after systemic application. The white bars in (A) and (B) indicate the

thickness of the outer nuclear layer. Red: glutamine synthetase, Müller cells spanning nearly

the entire thickness of the retina; blue: PNA, cone outer segments and endfeet; green: GFAP.

(A) untreated (B) DMSO i.p. (C) MNU i.p. 60 mg/kg BW (note that the ONL has nearly

vanished) (D) Glial reaction is indicated by GFAP expression in Müller cell processes (same

section as in (C)). (E) Rd10 mouse retina at the age of 50 days displaying total loss of

photoreceptors. OR: outer part of the retina; IR: inner part of the retina; OS: outer segments;

IS: inner segments; ONL: outer nuclear layer; OPL: outer plexiform layer; INL: inner nuclear

layer; IPL: inner plexiform layer; GCL: ganglion cell layer; NFL: nerve fiber layer.

Page 96: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

96

RESULTS

Figure 6. ERG after intravitreal MNU application. Examples of full-field ERGs recorded

before and one week after intravitreal (IVI) injection. (A) PBS (B) DMSO + PBS (C) 0.5

mg/kg BW MNU (D) 1 mg/kg BW MNU (E) 2 mg/kg BW MNU (F) 3 mg/kg BW MNU. (D)

– (F) A reduction of the amplitude of the ERG or a complete extinction is seen after

intravitreal injection of 1, 2, and 3 mg/kg BW MNU. In the right corner of (F) the ERG of the

contralateral, untreated right eye before and one week after treatment of the left eye

intravitreally with 3 mg/kg BW is exemplified illustrated to demonstrate that the treatment of

the left eye by intravitreal injection had no influence on photoreceptor function of the right

one. The Amplitudes of a-waves (G) and b-waves (H) are illustrated before and two weeks

after MNU application in different concentrations at a light intensity of 3 cds/m2 in the

scotopic ERG (n=3 for each concentration). In each diagram the bar designated with CL3

illustrates the values of the contralateral, right eyes of animals treated intravitreally with 3

mg/kg BW (left eye). There is no effect of intravitreal injection on the amplitude of a- and b-

wave in the ERG of the contralateral eye. Values represent mean ± SD.

Page 97: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

97

RESULTS

Figure 7. Retinal thickness evaluation using OCT upon intravitreal injection of MNU.

Retinal images and thickness obtained by sd-OCT measurements. Left panel. All images were

taken at the height of the papilla and displayed with pigment epithelium and photoreceptors to

the top. Examples of sd-OCT recordings before (left), one week after (middle), and two

weeks (right) after injection of PBS (A), DMSO + PBS (B), MNU 0.5 mg/kg BW (C), MNU

1 mg/kg BW (D), MNU 2 mg/kg BW (E), and MNU 3 mg/kg BW (F). (D) - (F) the red bars

mark the outer nuclear layer, which is significantly thinner one week after injection; the

Page 98: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

98

RESULTS

destruction of the ONL one week after application is reminiscent of the effects observed after

intraperitoneal application (compare to Figure 3C). (G) Retinal thickness measurements in the

height of the papilla based on sd-OCT data before, and two weeks after IVI injection of PBS,

DMSO/PBS and MNU. As already seen in Figure 5 CL3 illustrates the averaged retinal

thickness in the OCT of the contralateral, untreated eyes of animals injected intravitreally in

the left eye with 3 mg/kg BW MNU (highest concentration). Values represent mean ± SD (for

3 mg/kg BW MNU two weeks after application: ****p<0.0001 vs. the controls and the MNU-

treated animals before injection, one-way ANOVA with Bonferroni’s post hoc test). In (H)

the contralateral, corresponding untreated eye of the animal treated on the left eye

intravitreally with 3 mg/kg (F) two weeks after injection is exemplified illustrated. No

thickness differences or morphological changes in contrast to the eyes of control animals

could be observed (H).

Page 99: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

99

RESULTS

Figure 8. Confocal images of immunohistochemically stained vertical sections through

the retina two weeks after injection of MNU into the vitreous. The thickness of the ONL is

indicated by white bars. Stainings and abbreviations as in Figure 4. (A) Control injection of

PBS (B) Control injections of DMSO + PBS (C) 1 mg/kg BW MNU (D) 2 mg/kg BW MNU

(E) 3 mg/kg BW MNU. (C) - (E) Thickness of the ONL is progressively reduced. No

photoreceptors are left after injection of 3 mg/kg BW MNU (E). The bar diagrams in (B) and

(C) show the retinal thickness after different forms of treatment and in comparison to the

genetic model of the rd10 mouse at P50 (n=3). While in (B) the entire retinal thickness is

evaluated, in (C) only the thickness of the inner retinal layers is demonstrated, to evaluate the

possible effects of intravitreal MNU injection on these layers. (B) Retinal thickness decreases

Page 100: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

100

RESULTS

with rising intravitreal concentrations of MNU with the highest decrease after treatment with

3 mg/kg BW. The contralateral, untreated eye (CL3) is not affected regarding thickness. At

this concentration retinal thickness is comparable to the thickness after intraperitoneal (i.p.)

treatment with 60 mg/kg BW MNU or to the genetic model of the rd10 mouse. In (C) the

thickness measurements of the inner retinal layers demonstrate that intravitreal injections of

MNU have no influence on the thickness of these layers. The contralateral eyes of animals

treated by 3 mg/kg BW were not affected regarding thickness.

Page 101: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

101

RESULTS

Figure 9. Confocal images of immunohistochemically stained vertical sections through

the retina two weeks after injection of MNU into the vitreous. (A) Cartoon depicts the

injection site in the superior part of the eye. (B), (D), (F), (H): Stainings against recoverin

(green, entire photoreceptor), rhodopsin (blue, rod outer segment), and ion channel HCN1

(red, photoreceptor somata and inner segments, as well as processes in the inner retina). (C),

(E), (G), (I): Stainings against red/green opsin (green, cone outer segments in the superior

retina (C), (E)), blue opsin (red, cone outer segments in the inferior retina (G), (I)), PEA

(cones, blue). OR: outer half of the retina; IR: inner half of the retina. Photoreceptor

degeneration is more pronounced at the injection side. However, both, rods and cones, show

features of degeneration.

Page 102: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

102

RESULTS

Figure 10. Confocal images of immunohistochemically stained vertical sections through

the retina two weeks after injection of MNU. (A), (F), (K) wild type control. (B), (G), (L)

60 mg/kg BW MNU intraperitoneal. (C), (H), (M) 1 mg/kg BW MNU intravitreal. (D), (I),

(N) 2 mg/kg BW MNU intravitreal. (E), (J), (O) 3 mg/kg BW MNU intravitreal. Upper row:

antibodies against calbindin 28K yield strong label in horizontal cells (arrow) and weaker

Page 103: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

103

RESULTS

staining in amacrine cells. Middle row: PKCα is found in rod bipolar cells. Lower row:

calretinin is found in certain populations of amacrine cells. OPL: outer plexiform layer, IPL:

inner plexiform layer.

Page 104: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

104

GENERAL DISCUSSION

6 GENERAL DISCUSSION

It is important to study animal models with selective photoreceptor degeneration, which

resemble the human inherited disease Retinitis pigmentosa (RP); making reference to the

progress of degeneration, the different cell reactions and the work on possible treatment

strategies like surgical approaches. In rodents, many genetic models are available indicating

these conditions, e.g., the rd10 mouse. However, equivalent genetic models in larger animals

are intrinsically expensive and the duration until photoreceptor degeneration is completed

(TSO et al. 1997) is comparably long. Therefore, it would be very useful to have the

possibility to induce comparable outer retinal effects by administration of pharmaceuticals.

Photoreceptor degeneration could be induced in any wanted animal species over a faster

period of time. The general welfare of the experimental animals should not be affected by the

way of application. Iodoacetic acid (IAA) and N-methyl-N-nitrosourea (MNU) were used,

which are both known to lead to an outer retinal degeneration after systemic application

(GRAYMORE a. TANSLEY 1959, HERROLD 1967). However, after systemic treatment

toxic systemic side effects do occur. To this end, the - in this context “new” - technique of

intravitreal injections was introduced. The local administration of this injection technique was

hypothesized to induce photoreceptor degeneration similar to the effects obtained with

systemic application, but without affecting the contralateral eye as well as the general health

status of the animal.

Experiments were performed on wild type mice, in order to compare retinal effects to the

genetic model of the rd10 mouse, an important animal model revealing the requested and

offered conditions of retinal degeneration caused by a defect in the phosphodiesterase gene

(CHANG et al. 2007).

In the first study, the disease progression in the genetic model of the rd10 mouse was

characterised in comparison to the wild type mouse, which was examined as control.

Different in vivo and in vitro methods were used, and we demonstrated that the combination

of in vivo measurements by non-invasive tools like ERG, sd-OCT and fluorescein

angiography allow the precise description of the status of the retina during the degenerative

process. Our results were comparable to the findings of other researchers, e.g., the values of

latency (data not shown) and amplitudes of a-and b-wave in the ERG (CHANG et al. 2007,

Page 105: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

105

GENERAL DISCUSSION

SUGIMOTO et al 1997). In the fluorescein angiography, we could illustrate that there are no

significant differences between wild type and rd10 mice, although attenuated retinal vessels

were expected (BERSON 1993). The clinically used OCT was also proofed to be beneficial,

as we could clearly demonstrate that the thickness values obtained by OCT were comparable

to those obtained after histological analysis. However, retinal separation found by PENNESI

et al. (2012) at the age of 64 days, were only found in OCT measurements in vivo locally

determined in two animals (9 and 24 weeks of age). The differences in the results could be

probably explained by the use of different types of OCT devices (PENNESI et al. 2012).

However, in the histology the image of retinal separation was often observed, possibly

induced during the histological work-up. This may indicate that, although separation is not

always manifest in vivo, it can be more easily induced in the rd10 mouse in the histological

work-up, in contrast to the wild type (Study I).

Two protocols of histological work-up were chosen to enlarge the amount of data obtained.

HE-staining required sectioning of the whole eye into single slices. The advantage of this

method is the possibility to evaluate additional parts or tissues of the eye rather than the

neural retina, e.g., the lens. However, the disadvantage of this method is the frequent

observation of artifacts, like separation of the retina as seen in the rd10 mouse. In the

immunohistochemistry, only the retina was separated and cut into single slices. This method

allowed for the specific characterization of different cell classes of the retina, which can be

visualized in more detail using cell-type specific antibodies. The resulting data confirmed the

selective photoreceptor degeneration followed by retinal remodeling (CHANG et al. 2007,

PHILIPPS et al. 2010) (Study I).

In summary, the in vivo methods are very suitable to describe retinal changes and reduce the

number of animals sacrificed for histological techniques. The data obtained by these

measurements are in good accordance with the in vitro data (Study I).

After characterization of the rd10 mouse model, wild type mice were injected with two

different pharmaceuticals (iodoacetic acid (IAA; Study II) or N-methyl-N-nitrosourea (MNU,

Study III)) by different ways of application (systemic or intravitreal) in order to induce a

comparable outer retinal degeneration. Many publications, mainly in other species than mice,

demonstrate that, when both substances are systemically injected it leads to blindness causing

photoreceptor degeneration (LIANG et al. 2008, TSUBURA et al. 2010). The histologically

Page 106: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

106

GENERAL DISCUSSION

observed effect of both substances on photoreceptor survival is very similar to that presented

in the human disease of RP, when photoreceptors are completely degenerated (LIANG et al.

2008, TSUBURA et al. 2010). The systemic application of IAA, either given as an

intraperitoneal or as an intravenous injection, and the intraperitoneal application of MNU

consecutively served as positive controls in our experiments. However, both substances did

not only affect the retina after systemic treatment, they also led to a high reduction of the

welfare (Study II).

After systemic application of IAA (Study II), the welfare of the animals was sustainably

reduced for a couple of days, especially after intraperitoneal treatment. After intravenous

treatment, the highest level of mortality was observed. While the mortality rate after

intravenous IAA injections was reported to be 50% in rats (GRAYMORE et al. 1959) and

20% in rabbits (ORZALESI et al. 1970), the observed mortality in mice was 60%. IAA was

used and published in mice only once by SUGIMOTO et al. (1997), and the concentrations

(approximately 60 mg/kg BW) used in our experiments were chosen according to this

publication. We divided the 60 mg/kg BW into two dosages given within 3.5 hours

(GRAYMORE et al. 1959). This way of application did not lower the mortality, but IAA was

found to be more effective on the photoreceptor layer. The outer nuclear layer (ONL) was

reduced to 3-5 layers of somata. This effect was probably due to the maintaining of a critical

concentration for a longer and more sufficient period of time (GRAYMORE et al. 1959).

After systemic intraperitoneal application of IAA, no effects on the retina were seen, which

might be due to a minimally longer duration until passing the vasculature of the eye, in

contrast to intravenous application or because of a first pass effect in the liver (Study II).

In comparison to the systemic injection of IAA, animals receiving intraperitoneal application

of MNU (Study III) also showed a reduction of welfare for some hours after administration,

with the highest weight loss two days after injection. Nevertheless, all animals survived. The

ONL was nearly completely destroyed, comparable to the histological images of rd10 mice at

the age of P50. Inner retinal layers did not decrease in thickness. Horizontal cells and rod

bipolar cells were only slightly affected; this could be evaluated by the loss of most of their

dendrites; similar to findings described earlier (NAGAR et al. 2009) (Study III).

After systemic treatment with both of the substances, as well as our positive controls and

observation of their effects on the retinal structures and on the general welfare of the

Page 107: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

107

GENERAL DISCUSSION

experimental animals, we tried to induce a comparable retinal degeneration by intravitreal

injections of first IAA (Study II) and second MNU (Study III). Because of the small eye, the

proportionally large lens and the small vitreous space, the injection technique was difficult to

perform without affecting other structures of the eye. Therefore, the injections were

conducted using a 3D micromanipulator and a special nanoliter injection system. A volume of

2 µL was injected in the left eye of wild type mice. Previously, the method had been tested by

many test-injections (PBS, DMSO and PBS, fluorescein diluted with PBS), which were

carried out as negative controls and confirmed that the injection procedure was safe and

reproducible (Study II and Study III).

In contrast to the systemic treatment, which was conducted without anaesthesia, only a slight

weight loss was noticed after intravitreal injection, similar to that observed normally after an

anaesthetic intervention (Study II and Study III).

Intravitreally injected IAA (Study II) led to many side effects: dense cataracts, which

appeared five to seven days after injection, inflammation of different cell types in the eye

(e.g., iridocyclitis) and an exudation of blood cells into the aqueous humour. Because of the

later onset, the cataractous changes were not supposed to be the result of a lens injury during

the injection. In addition, they were not observed in control animals receiving injections other

than IAA. Due to the frequent cataract formation, retinal changes could not be evaluated in

the sd-OCT; the observed decrease of the waves in the ERG had to also be interpreted

carefully in consideration of the cataracts. After an experimental duration of two weeks, the

effects on the retina could be evaluated in immunohistochemistry. In contrast to the expected

effect of outer retinal degeneration, no selective photoreceptor degeneration was observed

upon intravitreal injection of IAA, but rather an overall thinning of the retina with a tendency

towards a more pronounced reduction of the inner retinal layers (Study II).

In summary, intravitreally injected IAA does not lead to a selective photoreceptor

degeneration. The injection led to cataract formation, inflammation and a thinning of the

retina, mainly based on a thinning of the inner retinal layers. In summary, the intravitreal

injection of IAA is no useful method to induce unilateral photoreceptor degeneration (Study

II).

One week after injection, animals treated intravitreally with MNU (1, 2 or 3 mg/kg BW

MNU; Study III) showed, correlated to the concentration, similar changes and cell debris in

Page 108: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

108

GENERAL DISCUSSION

the outer nuclear layer in the OCT as animals which were examined one week after

intraperitoneal treatment. In the groups of animals treated with 1 and 2 mg/kg BW MNU the

values of a- and b-wave in the ERG were either diminished or even extinguished. Histology

revealed that the ONL had not completely disappeared. Additionally, we pointed out that the

degree of photoreceptor degeneration depended on the distance to the injection site. Retinal

areas of treated eyes, which were not near to the injection side and were thus less affected,

comprised both rods and cones. In the group of animals treated intravitreally with 3 mg/kg

BW we observed a degeneration of all photoreceptors and somata throughout the entire eye in

the OCT. No response could be observed in the ERG. In the immunohistochemistry, we

verified the OCT findings of no surviving photoreceptors (Study III). The thickness of inner

retinal layers remained unaffected. This described histological picture was similar to those

obtained after systemic MNU treatment or to those retinal histological pictures of the rd10

mouse at P50. However, horizontal cells and rod bipolar cells were obviously more affected

than after intraperitoneal application. In those regions, where photoreceptors were completely

lost, horizontal cell somata and processes had also disappeared (Study III). In contrast, three

months after systemic treatment, only a loss of dendritic processes of horizontal cells is

described (NAGAR et al. 2009). In the genetic model of the rd10 mouse at the age of 9

months, a loss of horizontal cell somata of approximately 29% was described (GARGINI et

al. 2007).

Apart from horizontal cells, the rod bipolar cells also seemed to be affected and to be

disorganized after an intravitreal injection of 3 mg/kg BW MNU (Study III). While in rd10

mice up to 45 days of age, no changes in the morphology of these cells were found, the

number of rod bipolar cells decreased by 20% between 1.5 and 3.5 months of age (GARGINI

et al. 2007).

One explanation, as to why intravitreally injected MNU affected the horizontal and rod

bipolar cells would be that MNU, after the death of photoreceptors, might become toxic to

horizontal cells and rod bipolar cells in higher concentrations (Study III). Secondly, the more

rapid loss of photoreceptors after intravitreal injection, in contrast to the genetic model or the

systemic treatment might have a more pronounced effect on the survival of postsynaptic cells

(Study III).

Page 109: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

109

GENERAL DISCUSSION

However, in all intravitreally treated animals the thickness of the inner retinal layers did not

significantly decrease. And most importantly, ganglion cells morphologically seemed not to

be affected after intravitreal MNU treatment.

In summary, intravitreal application of MNU led to unilateral selective photoreceptor

degeneration comparable to a complete loss of photoreceptors after systemic treatment or in

the later stages of the genetic model of the rd10 mouse. The well-being of the animal was not

more affected than after a normal anaesthetic intervention. Additionally, the contralateral eye

was not affected. We suggested the use of MNU injections into the vitreous of animals to

induce unilateral photoreceptor degeneration, while maintaining an intraindividual control eye

(Study III).

Page 110: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

110

SUMMARY

7 SUMMARY

Sarah Rösch

Intravitreal injections in mice as a technique to induce unilateral photoreceptor

degeneration in comparison to the genetic model of the rd10 mouse

Inherited blindness causing diseases in humans are frequent and often issue of new

therapeutic strategies in research. To test these strategies, but also for a better understanding

of the pathological mechanisms, animal models that mimic pathohistological changes in the

human retina are indispensable.

For Retinitis pigmentosa, a disease with selective photoreceptor degeneration and remaining

inner retinal layers, genetic animal models in rodents allow the characterisation of the process

of degeneration. However, for the testing of visual prostheses to restore visual function or

related surgical methods, the eyes of these genetic animal models are too small. For these

experiments, animal species such as the rabbit, the cat or the pig are required, but such genetic

models for larger animals are intrinsically expensive and have a slow disease progression.

However, photoreceptor degeneration can also be induced by systemic treatment with

iodoacetic acid (IAA) and N-methyl-N-nitrosourea (MNU), both leading to substantial side

effects. To induce the same retinal changes as seen in the genetic model without affecting the

welfare of the animals, experiments were performed using the - in this context “new” -

technique of intravitreal injections. The mouse was used as animal species in these

experiments in order to compare the results to the available genetic model of the retinally

degenerated rd10 mouse.

Firstly, the process of photoreceptor degeneration and its characteristics in the genetic model

of the rd10 mouse was observed and evaluated by clinical tools (OCT, ERG, FA, histology)

in contrast to the healthy wild type mouse. Secondly, effects of systemic and intravitreal

treatment using either IAA or MNU were compared. Systemic treatment with IAA or MNU

led to outer retinal degeneration; however, the systemic application of either substance

Page 111: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

111

SUMMARY

resulted in a high reduction of animal welfare or even in mortality (in the case of IAA) by

systemic side effects.

Upon the third approach, utilizing unilateral intravitreal injection of MNU or IAA under

anaesthesia, no reduction of animal welfare was observed and no effects were observed in the

uninjected eye. The intravitreal injection of IAA led to cataract formation and inflammatory

reactions of the whole eye. Retinal thickness was reduced mainly due to thinning of the inner

retinal layers, rather than the photoreceptor layer. Intravitreal application of MNU led to

photoreceptor degeneration similar to the effects seen after systemic treatment and resembling

the degeneration in the genetic model.

To conclude, the intravitreal injection of MNU is a suitable model to induce outer retinal

degeneration, without affecting the general health status of the animal and with preservation

of an intraindividual control eye avoiding sudden complete blindness.

Page 112: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

112

ZUSAMMENFASSUNG

8 ZUSAMMENFASSUNG

Sarah Rösch

Technik der intravitrealen Injektion bei der Maus zur Induktion einer einseitigen

Photorezeptordegeneration im Vergleich zum genetischen Modell der rd10 Maus

Genetisch bedingte, unheilbare Netzhauterkrankungen wie Retinitis Pigmentosa (RP) führen

zu einer Degeneration der Photorezeptoren und somit final zur Erblindung. Aufgrund ihres

häufigen Vorkommens sind sie zunehmend Thema neuer Forschungsansätze auf der Suche

nach effektiven Therapiemöglichkeiten. Um sowohl diese neuen therapeutischen Ansätze

testen zu können, aber auch um die pathohistologischen Vorgänge besser verstehen zu

können, sind Tiermodelle unverzichtbar, die vergleichbare Veränderungen aufweisen.

Genetisch veränderte Tiermodelle in Nagern, die die Verhältnisse wie bei RP widerspiegeln,

sind verfügbar und erlauben eine Charakterisierung der selektiven Photorezeptordegeneration

und die Evaluierung der Auswirkungen auf die verbleibenden, intakten inneren Zellen der

Netzhaut. Für das Austesten von Sehprothesen, die eine visuelle Funktion ansatzweise

wiederherstellen sollen, oder aber für vergleichbare chirurgische therapeutische Ansätze, sind

die Augen dieser Tierspezies zu klein. Man würde hierzu Tierarten benötigen, wie z.B. das

Kaninchen, die Katze oder das Schwein, die größere Augen besitzen. Genetisch veränderte

Tiermodelle dieser Tierarten mit einer gleichartigen Erblindung sind jedoch sehr schwer zu

beziehen, sehr teuer und zeigen eine nur sehr langsame Photorezeptordegeneration.

Allerdings kann eine Photorezeptordegeneration auch durch eine systemische Verabreichung

von pharmakologischen Wirkstoffen erreicht werden, wie z.B. mit Jodessigsäure (Iodoacetic

Acid, IAA) oder n-Methyl-n-Nitrosoharnstoff (N-methyl-N-nitrosourea, MNU). Die

systemische Gabe beider Substanzen führt jedoch zu starken Beeinträchtigungen der

Tiergesundheit und des Wohlbefindens. Um die gleichen netzhautspezifischen

Veränderungen zu induzieren, ohne jedoch das Allgemeinwohl des Tieres zu beeinflussen,

wurde in den dargestellten Experimenten IAA und MNU mit der in diesem Zusammenhang

„neuen“ Methode der intravitrealen Injektion verabreicht. Als Tierspezies wurde die Maus

Page 113: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

113

ZUSAMMENFASSUNG

gewählt, da hier das genetische Modell der rd10 Maus mit der erwünschten Form der

Photorezeptordegeneration als Vergleich herangezogen werden konnte.

Zuerst wurde die Netzhautdegeneration in der rd10 Maus mittels ERG, OCT und die

Histologie (Immunhistochemie und HE-Färbungen) im Vergleich zu der gesunden Wildtyp

Maus beobachtet und dargestellt. Anschließend wurden die Effekte durch die systemische und

intravitreale Applikation beider Substanzen analysiert. Zwar führte die systemische

Applikation als Positiv-Kontrolle wie erwartet zu einer äußeren Netzhautdegeneration, war

jedoch mit einer starken Beeinflussung des Tierwohls oder sogar Mortalität (im Fall von IAA)

verbunden.

Die unter Anästhesie durchgeführte unilaterale intravitreale Injektion beider Substanzen

führte zu keiner Reduktion des Tierwohls und zu keinerlei Effekten im zweiten,

unbehandelten Auge. Intravitreal verabreichtes IAA führte in erster Linie zu einer Katarakt

und zu Entzündungsreaktionen im Auge. Zwar war nach der Injektion die Netzhautdicke

reduziert, dies basierte jedoch vor allem auf einer Reduktion der inneren Netzhautschichten.

Die intravitreale Injektion von MNU führte zu einer Photorezeptordegeneration vergleichbar

mit der systemischen Applikation oder dem genetischen Modell.

Damit lässt sich schlussfolgern, dass die intravitreale Injektion von MNU ein gutes und

notwendiges Modell der induzierbaren Photorezeptordegeneration darstellt, in dem das

allgemeine Wohl der Versuchstiere nicht negativ beeinflusst und das kontralaterale Auge

erhalten wird und somit als intraindividuelles Kontrollauge dienen kann. Eine plötzliche,

komplette Erblindung nach Applikation wird somit vermieden.

Page 114: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

114

REFERENCES

9 REFERENCES

AHL, A.S. (1986):

The role of vibrissae in behavior: a status review.

Vet. Res. Commun. 10, 245-268

AHUJA, A., P. AHUJA-JENSEN, L.E. JOHNSON, A.R. CAFFÉ, M. ABRAHAMSON, P.A.

EKSTRÖM, T. VAN VEEN (2008):

Rd1 Mouse retina shows an imbalance in the activity of cysteine protease cathepsins and their

endogenous inhibitor cystatin C.

Invest. Ophthalmol. Vis. Sci. 49, 1089-1096

ALFARO, Y., G. DELGADO, A. CÀBAREZ, B. ANGUIANO, C. ACEVES (2013):

Iodine and doxorubicin, a good combination for mammary cancer treatment: antineoplastic

adjuvancy, chemoresistance inhibition, and cardioprotection.

Mol. Cancer. Doi:10.1186/1476-4598-12-45

BAID, R., A.K. UPADHYAY, T. SHINOHARA, U.B. KOMPELLA (2013):

Biosynthesis, Characterization, and Efficacy in Retinal Degenerative Disease of Lens

Epithelium-derived Growth Factor Fragment (LEDGF1-326), a Novel Therapeutic Protein.

J. Biol. Chem. 288, 17372-17383

BERSON, E.L. (1993):

Retinitis pigmentosa: The Friedenwald Lecture.

Invest. Ophthalmol. Vis. Sci. 34, 1659-1676

BERUFSVERBAND DER AUGENÄRZTE DEUTSCHLANDS E.V., DEUTSCHE

OPHTHALMOLOGISCHE GESELLSCHAFT (DOG) (2010):

Augenärzte informieren: Intravitreale Medikamenteneingabe, operative Medikamenten-

eingabe in das Auge.

Düsseldorf und München

CHANG, B., N.L. HAWES, R.E. HURD, M.T. DAVISSON, S. NUSINOWITZ, J.R.

HECKENLIVELY (2002):

Retinal degeneration mutants in the mouse.

Vision Res. 42, 517-525

CHANG, B., N.L. HAWES, M.T. PARDUE, A.M. GERMAN, R.E. HURD, M.T.

DAVISSON, S. NUSINOWITZ, K. RENGARAJAN, A.P. BOYD, S.S. STARR, R.C.

CHAUDHURY, J.M. NICKERSON, J.R. HECKENLIVELY, J.H. BOATRIGHT (2007):

Two mouse retinal degenerations caused by missense mutations in the beta-subunit of rod

cGMP phosphodiesterase gene.

Vision Res. 47, 624-633

Page 115: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

115

REFERENCES

CHEN, M., K. WANG, B. LIN (2012):

Development and degeneration of cone bipolar cells are independent of cone photoreceptors

in a mouse model of retinitis pigmentosa.

PLoS ONE 7, e44036

CHRENEK, M.A., N. DALAL, C. GARDNER, H. GROSSNIKLAUS, Y. JIANG, J.H.

BOATRIGHT, J.M. NICKERSON (2012):

Analysis of the RPE sheet in the rd10 retinal degeneration model.

Adv. Exp. Med. Biol. 723, 641-647

CIBIS, P.A., M. CONSTANT, A. PRIBYL, B. BECKER (1957):

Ocular lesions produced by iodoacetate.

AMA Arch. Ophthalmol. 57, 508-519

DELYFER, M.N., V. FORSTER, N. NEVEUX, S. PICAUD, T. LÉVEILLARD, J.A. SAHEL

(2005):

Evidence for glutamate-mediated excitotoxic mechanisms during photoreceptor degeneration

in the rd1 mouse retina.

Mol. Vis. 11, 688-696

DUREAU, P., J. JEANNY, B. CLERC, J. DUFIER, Y. COURTOIS (1996):

Long Term Light-Induced Retinal Degeneration in the Miniature Pig.

Mol. Vis. 2, 7

EISENFELD, A.J., A.H. BUNT-MILAM, P.V. SARTHY (1984):

Müller cell Expression of Glial Fibrillary Acidic Protein after Genetic and Experimental

Photoreceptor Degeneration in the Rat Retina.

Invest. Ophthalmol. Vis. Sci. 25, 1321-1328

FEUCHT, M., T. LAUBE, N. BORNFELD, P. WALTER, M. VELIKAY-PAREL, R.

HORNIG, G. RICHARD (2005):

Entwicklung einer epiretinalen Prothese zur Stimulation der humanen Netzhaut.

Ophthalmologe 102, 688-691

FLETCHER, E.L., M. KALLONIATIS (1997):

Neurochemical development of the degenerating rat retina.

J. Comp. Neurol. 388, 1-22

GARGINI, C., E. TERZIBASI, F. MAZZONI, E. STRETTOI (2007):

Retinal Organization in the retinal degeneration 10 (rd10) Mutant Mouse: a morphological

and ERG Study.

J. Comp. Neurol. 500, 222-238

GIBSON, R., E.L. FLETCHER, A.J. VINGRYS, Y. ZHU, K.A. VESSEY, M.

KALLONIATIS (2013):

Functional and neurochemical development in the normal and degeneration mouse retina.

J. Comp. Neurol. 521, 1251-1267

Page 116: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

116

REFERENCES

GRAYMORE, C., K. TANSLEY (1959):

Iodoacetate poisoning of the rat retina- II Glycolysis in the poisoned retina.

Brit. J. Ophthal. 43, 486-493

HARADA, T., C. HARADA, M. SEKIGUCHI, K. WADA (1998):

Light-induced retinal degeneration suppresses developmental progression of flip-to-flop

alternative splicing in GluR1.

J. Neurosci. 18, 3336-3343

HART, A.W., L. MCKIE, J.E. MORGAN, P. GAUTIER, K. WEST, I.J. JACKSON, S.H.

CROSS (2005):

Genotype-phenotype correlation of mouse pde6b mutations.

Invest. Ophthalmol. Vis. Sci. 46, 3443-3450

HARTONG, D.T., E.L. BERSON, T.P. DRYJA (2006):

Retinitis pigmentosa.

Lancet. 368, 1795-1809

HEINEMANN, A., K. MEICHNER (2011):

Sicherer Umgang mit Zytostatika in der Veterinärmedizin.

Prakt. Tierarzt 12, 1114-1118

HERROLD, K.M. (1967):

Pigmentary degeneration of the retina induced by N-methyl-N-nitrosourea. An experimental

study in Syrian hamsters.

Arch. Ophthalmol. 78, 650-653

HESS, H.H., D.A. NEWSOME, J.J. KNAPKA, G.E. WESTNEY (1982):

Slitlamp assessment of age of onset and incidence of cataracts in pink-eyed, tan-hooded

retinal dystrophic rats.

Curr. Eye Res. 2, 265-269

HICKS, D., J. SABEL (1999):

The Implications of Rod-Dependent Cone Survival for Basic and Clinical Research.

Invest. Ophthalmol. Vis. Sci. 40, 3071-3074

HIROTA, R., M. KONDO, S. UENO, T. SAKAI, T. KOYASU, H. TERASAKI (2012):

Photoreceptor and post-photoreceptoral contributions to photopic ERG a-wave in rhodopsin

P347L transgenic rabbits.

Invest. Ophthalmol. Vis. Sci. 53, 1467-1472

JONES, B.W., M. KONDO, H. TERASAKI, C.B. WATT, K. RAPP, J. ANDERSON, Y.

LIN, M.V. SHAW, J.H. YANG, R.E. MARC (2011):

Retinal remodeling in the TG P347L rabbit, a large-eye model of retinal degeneration.

J. Comp. Neurol. 519, 2713-2733

Page 117: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

117

REFERENCES

KONDO, M., T. SAKAI, K. KOMEIMA, Y. KURIMOTO, S. UENO, Y. NISHIZAWA, J.

USUKURA, T. FUJIKADO, Y. TANO, H. TERASAKI (2009):

Generation of a Transgenic Rabbit Model of Retinal Degeneration.

Invest. Ophthalmol. Vis. Sci. 50, 1371-1377

KRISHNA, V.R., K. R. ALEXANDER, N. S. PEACHEY (2002):

Temporal Properties of the Mouse Cone Electroretinogram

J. Neurophysiol. 87, 42-48

LIANG, L., Y. KATAGIRI; L.M. FRANCO, Y. YAMAUCHI, V. ENZMANN, H.J.

KAPLAN, J.H. SANDELL (2008):

Long-term cellular and regional specificity of th photoreceptor toxin, iodoacetic acid (IAA),

in the rabbit retina.

Vis. Neuroscience 25, 167-177

MARMOR, M.F., A.B. FULTON, G.E. HOLDER, Y. MIYAKE, M. BRIGELL, M. BACH

(2008):

ISCEV Standard for full-field clinical electroretinography (2008 update).

Doc. Ophthalmol. 118, 69-77

MATARUGA, A., E. KREMMER, F. MÜLLER (2007):

Type 3a and type 3b OFF cone bipolar cells provide for the alternative rod pathway in the

mouse retina.

J Comp Neurol. 502: 1123-1137

MATSUMOTO, M., T. MORITA, S. HIRAYAMA, S. UGA, K. SHIMIZU (2002):

Morphological Analysis of Iodoacetic Acid-Induced Cataract in the Rat.

Ophthalmic Res. 34, 119-127

MATUK, Y. (1984):

Retinitis pigmentosa and retinal degeneration in animals: a review.

Can. J. Biochem. Cell Biol. 62, 535-546

MCCORMICK, D.L., K.V. RAO, L. DOOLEY, V.E. STEELE, R.A. LUBET, G.J.

KELLOFF, M.C. BOSLAND (1998):

Influence of N-methyl-N-nitrosourea, testosterone, and N-(4-hydroxyphenyl)-all-trans-

retinamide on prostate cancer induction in Wistar-Unilever rats.

Cancer Res. 58, 3282-3288

MENOTTI-RAYMOND, M., V. DAVID, A. SCHÄFFER, R. STEPHENS, D. WELLS, R.

KUMAR-SINGH, S. O´BRIEN, K. NARFSTRÖM (2007):

Mutation in CEP 290 Discovered for Cat Model of Human Retinal Degeneration.

J. Hered. 98, 211-220

MENZEL-SEVERING, J., T. LAUBE, C. BROCKMANN, N. BORNFELD, W. MOKWA,

B. MAZINANI, P. WALTER, G. ROESSLER (2012):

Page 118: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

118

REFERENCES

Implantation and explantation of an active epiretinal visual prosthesis: 2-year follow-up data

form the EPIRET3 prospective clinical trial.

Eye 26, 501-509

MURAOKA, Y., H.O. IKEDA, N. NAKANO, M. HANGAI, Y. TODA, K. OKAMOTO-

FURUTA, H. KOHDA, M. KONDO, H. TERASAKI, A. KAKIZUKA, N. YOSHIMURA

(2012):

Real-time imaging of rabbit retina with retinal degeneration by using sprectral-domain optical

coherence tomography.

PLoS ONE 7, e36135

NAGAR, S., V. KRISHNAMOORTHY, P. CHERUKURI, V. JAIN, N.K. DHINGRA

(2009):

Early remodeling in an inducible animal model of retinal degeneration.

Neuroscience 160, 517-529

NAMBU, H., K. YUGE, M. NAKAJIMA, N. SHIKATA, K. TAKAHASHI, H. MIKI, M.

UYAMA, A. TSUBURA (1997):

Morphologic characteristics of N-methyl-N-nitrosourea-induced retinal degeneration in

C57BL mice.

Pathol. Int. 47, 377-383

NATIONAL INSTITUTE FOR HEALTH RESEARCH (NIHR) NATIONAL HORIZONTM

SCANNING CENTRE RESEARCH PROGRAMME (2011):

Second Sight Medical Products Argus IITM

Retinal Prosthesis System for peripheral retinal

degeneration.

University of Birmingham, United Kingdom

NICKEL, R., A. SCHUMMER, E. SEIFERLE (2004):

Lehrbuch der Anatomie der Haustiere Band 4

Paul Parey, Berlin und Hamburg, 4. Auflage, S. 405-444

NISHIDA K., M. KAMEI, M. KONDO, H. SAKAGUCHI, M. SUZUKI, T. FUJIKADO, Y.

TANO (2010):

Efficacy of suprachoroidal-transretinal stimulation in a rabbit model of retinal degeneration.

Invest. Ophthalmol. Vis. Sci. 51, 2263-2268

NOEL, J., J.P. FERNANDEZ DE CASTRO, P. J. DEMARCO JR., L.M. FRANCO, W.

WANG, E.V. VUKMANIC, X. PENG, J.H. SANDELL, P.A. SCOTT, H.J. KAPLAN, M.A.

MCCALL (2012):

Iodoacetic acid, but not sodium iodate, creates an inducible swine model of photoreceptor

damage.

Exptl. Eye Res. 97, 137-147

NOELL, W.K. (1953):

Impairment of visual cell structure by iodoacetate.

J. Cell. Physiol. 40, 25-55

Page 119: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

119

REFERENCES

NOELL, W.K. (1953):

Experimentally induced toxic effects on structure and function of visual cells and pigment

epithelium.

Am. J. Ophthalmol. 36, 103-116

ORZALESI, N., G.A. CALABRIA, A. GRIGNOLO (1970):

Experimental Degeneration of the Rabbit Retina induced by Iodoacetic Acid.

Exptl. Eye Res. 9, 246-253

PANG, J.J., S.L. BOYE, A. KUMAR, A. DINCULESCU, W. DENG, J. LI, Q. LI, A. RANI,

T.C. FOSTER, B. CHANG, N.L. HAWES, J.H. BOATRIGHT, W.W. HAUSWIRTH (2008):

AVV-mediated gene therapy for retinal degeneration in the rd10 mouse containing a recessive

PDEbeta mutation.

Invest. Ophthalmol. Vis. Sci. 49, 4278-4283

PERCHE, O., M. DOLY, I. RANCHON-COLE (2008):

Transient protective effect of caspase inhibitors in RCS rat.

Exp. Eye Res. 86, 519-527

PENNESI, M.E., K.V. MICHAELS, S.S. MAGEE, A. MARICLE, S.P. DAVIN, A.G.

GARG, M.J. GALE, D.C. TU, Y. WEN, L.R. ERKER, P.J. FRANCIS (2012):

Long-term characterization of retinal degeneration in rd1 and rd10 mice using spectral

domain optical coherence tomography.

Invest. Ophthalmol. Vis. Sci. 53, 4644-4656.

PHILLIPS, M.J., D.C. OTTESON, D.M. SHERRY (2010):

Progression of neuronal and synaptic remodeling in the rd10 mouse model of retinitis

pigmentosa.

J. Comp. Neurol. 518, 2071-2089.

RAPP, L.M., T.P. WILLIAMS (1980):

The role of ocular pigmentation in protecting against retinal light damage.

Vis. Res. 20, 1127-1131

RICHTLINIE 2010/63/EU DES EUROPÄISCHEN PARLAMENTS UND DES RATES

(2010):

Zum Schutz der für wissenschaftliche Zwecke verwendeten Tiere

ROESSLER, G., T. LAUBE, C. BROCKMANN, T. KIRSCHKAMP, B. MAZINANI, M.

GOERTZ, C. KOCH, I. KIRSCH, B. SELLHAUS, H.K. TRIEU, J. WEIS, N. BORNFELD,

H. RÖTHGEN, A. MESSNER, W. MOKWA, P. WALTER (2009):

Implantation and Explantation of a Wireless Epiretinal Retina Implant Device: Observations

during the EPIRET3 Prospective Clinical Trial.

Invest. Ophthalmol. Vis. Sci. 50, 3003-3008

Page 120: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

120

REFERENCES

SAMARDZIJA, M., H. WARIWODA, C. IMSAND, P. HUBER, S.R. HEYNEN, A.

GUBLER, C. GRIMM (2012):

Activation of survival pathways in the degenerating retina of rd10 mice.

Exp. Eye Res. 99, 17-26

SCHREIBER, D., W. JÄNISCH, R. WARZOK, H. TAUSCH (1969):

Induction of brain and spinal cord tumors in rabbits with N-methyl-N-nitrosourea.

Z Gesamte Exp. Med. 150, 76-86

SCOTT, P.A., H.J. KAPLAN, J.H. SANDELL (2011):

Anatomical evidence of photoreceptor degeneration induced by iodoacetic acid in the porcine

eye.

Exp. Eye Res. 93, 513-527

SHIVELY, J.N., R.D. PHEMISTER, G.P. EPLING, R. JENSEN (1970):

Pathogenesis of radiation-induced retinal dysplasia.

Invest. Ophthalmol. Vis. Sci. 9, 888-900

SIGMA-ALDRICH® (2013)

N-nitroso-N-methylurea Isopac®

Saint Louis, MO 63103, USA

SMITH, C.P., V. TAYLOR (1995):

Environmental Enrichment Information Resources for Laboratory Animals: 1965-1995:

Birds, Cats, Dogs, Farm animals, Ferrets, Rabbits, and Rodents.

AWIC Resource No. 2. U.S. Department of Agriculture

SUGIMOTO, S., M. IMAWAKA, R. IMAI, K. KANAMARU, T. ITO, S. SASAKI, T.

ANDO, T. SAIJO, S. SATO (1997):

A procedure for electroretinogram (ERG) recording in mice - effect of monoiodoacetic acid

on the ERG in pigmented mice.

J. Toxicol. Sci. 22, 315-325

TERAI, N., SANDNER, D., KISSNER, A., PILLUNAT, L.E. (2011):

Siderosis bulbi nach intraokularem Metallfremdkörper.

Ophthalmologe 108, 60-63

TIERI, O., L. VECCHIONE (1963):

Monolateral cataract provoked with Iodoacetic acid.

Acta ophthalmol. 41, 205-212

TIERSCHUTZGESETZ (2013)

Fünfter Abschnitt: Tierversuche § 7a

Page 121: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

121

REFERENCES

TSO, M., W. LI, C. ZHANG, T. LAM, Y. HAO, R. PETTERS, F. WONG (1997):

A pathologic study of degeneration of the rod and cone populations of the rhodopsin

Pro347Leu transgenic pigs.

Trans. Am. Ophthalmol. Soc. 95, 479-483

TSUBURA, A., K. YOSHIZAWA, K. KIUCHI, K. MORIGUCHI (2003):

N-Methyl-N-nitrosourea-induced Retinal Degeneration in Animals.

Acta Histochem. Cytochem. 36, 263-270

TSUBURA, A., K. YOSHIZAWA, M. KUWATA, N. UEHARA (2010):

Animal models for retinitis pigmentosa induced by MNU; disease progression, mechanisms

and therapeutic trials.

Histol. Histopathol. 25, 933-944

TSURUMA, K., M. YAMAUCHI, Y. INOKUCHI, S. SUGITANI, M. SHIMAZAWA, H.

HARA (2012):

Role of Oxidative Stress in Retinal Photoreceptor Cell Death in N-Methyl-N-nitrosourea-

Treated Mice.

J. Pharmacol. Sci. 118, 351-362

TUNTIVANICH, N., S.J. PITTLER, A.J. FISCHER, G. OMAR, M. KIUPEL, A. WEBER, S.

YAO, J.P. STEIBEL, N.W. KHAN, S.M. PETERSEN-JONES (2009):

Characterization of a Canine Model of Autosomal Recessive Retinitis Pigmentosa due to a

PDE6A Mutation.

Invest. Ophthalmol. Vis. Sci. 50, 801-813

VON ENGELHARDT, W., G. BREVES (2004):

Physiologie der Haustiere

Enke, Stuttgart, 2. Auflage

WALDE, I., B. NELL, E.H. SCHÄFFER, R.G. KÖSTLIN (2008):

Augenheilkunde – Lehrbuch und Atlas – Hund, Katze, Kaninchen und Meerschweinchen

Schattauer, Stuttgart und New York, 3. Auflage

WALTER, P. (2005):

Elektronische Sehprothesen

Klin. Monatsbl. Augenheilkd. 222: 471-479

Georg Thieme Verlag KG Stuttgart - New York

WANG, W., J.F. DE CASTRO, E. VUKMANIC, L. ZHOU, D. EMERY, P.J. DEMARCO,

H.J. KAPLAN, D.C. DEAN (2011):

Selective Rod Degeneration and Partial Cone Inactivation Characterize an Iodoacetic Acid

Model of Swine Retinal Degeneration.

Invest. Ophthalmol. Vis. Sci. 52, 7917-7923

Page 122: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

122

REFERENCES

WANG, Z., K. LAM, T. LAM, M. ON MAN TSO (1998):

Iron-induced apoptosis in the photoreceptor cells of rats.

Invest. Ophthalmol. Vis. Sci. 39, 631-633

WINKLER, B.S., M.W. SAUER, C.A. STARNES (2003):

Modulation of the Pasteur effect in retinal cells: implications for understanding compensatory

metabolic mechanisms.

Exp. Eye Res. 76, 715-723

YAMAUCHI, Y., T. AGAWA, R. TSUKAHARA, K. KIMURA, N. YAMAKAWA, M.

MIURA, H. GOTO (2011):

Correlation between high-resolution optical coherence tomography (OCT) images and

histopathology in an iodoacetic acid-induced model of retinal degeneration in rabbits.

Br. J. Ophthalmol. 95, 1157-1160

YAMAUCHI, Y., K. KIMURA, T. AGAWA, R. TSUKAHARA, M. MISHIMA, N.

YAMAKAWA, H. GOTO (2011):

Correlation between high-resolution optical coherence tomography (OCT) images and

histopathology in an N-methyl-N-nitrosourea-induced retinal degeneration rat model.

Br. J. Ophthalmol. 95, 1161-1165

YOSHIZAWA, K., J. YANG, H. SENZAKI, Y. UEMURA, Y. KIYOZUKA, N. SHIKATA,

Y. OISHI, H. MIKI, A. TSUBURA (2000):

Caspase-3 inhibitor rescues N-methyl-N-nitrosourea-induced retinal degeneration in Sprague-

Dawley rats.

Exp. Eye Res. 71, 629-635

Page 123: University of Veterinary Medicine Hannover · Sarah Rösch1,2, Sandra Johnen1, Frank Müller3, Christiane Pfarrer2, Peter Walter1 1Department of Ophthalmology, RWTH Aachen University,

123

ACKNOWLEDGEMENTS

10 ACKNOWLEDGEMENTS

I would like to thank my supervisors Prof. Dr. Christiane Pfarrer and Prof. Dr. Peter Walter

for the great opportunity to work on this topic and project, and for the possibility to learn so

much about urgent themes in the complex “research”. I express my gratitude to Prof. Dr. Peter

Walter for giving me an idea about how important the connection between “research and

results” is for sufferers of different diseases.

I would like to thank the second referee PD PhD Veronika Stein for reviewing this thesis.

I would also like to thank very much the people working at the department of ophthalmology

in Aachen for their support and the very pleasant working atmosphere.

Furthermore, I would like to thank Prof. Dr. Frank Müller very much for the great support and

inspiring discussions about the project and concerning questions, and for the possibility to

prepare the immunohistochemistry in his laboratory and the use of his equipment. Also I

would like to thank Christoph Aretzweiler very much, working at the same institute for his

support in the preparation of the retinae and in immunohistological stainings of the eyes and

the time he took to explain working protocols. I learned very much.

I would especially like to thank Prof. Dr. R. Tolba and Dr. K. Scherer of the Institute of

Laboratory Animal Science in Aachen, because without their cooperativeness, their support

and their encouragement, the experiments, especially with the substance MNU, would not

have taken place. I really appreciated the opportunity to perform the experiments in an

isolated laboratory of the Institute of Laboratory Animal Science. Moreover, I would like to

thank Dr. K. Scherer very much for the enlightening talks about different themes of the

laboratory animal science.

Last but not least, I would like to thank my parents and most especially my dog Toto, for

showing an inexhaustible amount of understanding. Also, I want to thank my friends and

former working colleagues in Jülich, Aachen, Mönchengladbach and Hannover (“D”) for their

support, encouragement, helping words and/ or just for being there…

Without you, this academic journey would have been much more difficult…