106

ISBN 978-3-86345-286-5 - elib.tiho-hannover.de · ISBN 978-3-86345-286-5 Verlag: Deutsche Veterinärmedizinische Gesellschaft Service GmbH 35392 Gießen · Friedrichstraße 17 ·

  • Upload
    others

  • View
    28

  • Download
    0

Embed Size (px)

Citation preview

ISBN 978-3-86345-286-5

Verlag: Deutsche Veterinärmedizinische Gesellschaft Service GmbH35392 Gießen · Friedrichstraße 17 · Tel. 0641 / 24466 · Fax: 0641 / 25375

E-Mail: [email protected] · Internet: www.dvg.de

Mu

ham

mad

Nad

eem

Han

no

ver

2015

Bibliografische Informationen der Deutschen Bibliothek

Die Deutsche Bibliothek verzeichnet diese Publikation in der

Deutschen Nationalbibliografie;

Detaillierte bibliografische Daten sind im Internet über http://dnb.ddb.de abrufbar.

1. Auflage 2015

© 2015 by Verlag: Deutsche Veterinärmedizinische Gesellschaft Service GmbH,

Gießen

Printed in Germany

ISBN 978-3-86345-286-5

Verlag: DVG Service GmbH

Friedrichstraße 17

35392 Gießen

0641/24466

[email protected]

www.dvg.de

University of Veterinary Medicine Hannover

Department of Pathology

Susceptibility of goats to the BSE agent with special emphasis on the neuropathogenesis

THESIS

Submitted in partial fulfillment of the requirements for the degree

DOCTOR OF PHILOSOPHY

(PhD)

Awarded by the University of Veterinary Medicine Hannover

By

Muhammad Nadeem

Born in Faisalabad, Punjab/Pakistan

Hannover, Germany 2015

Supervisor: Prof. Dr. Wolfgang Baumgärtner

Supervision Group: Prof. Dr. Wolfgang Baumgärtner

Prof. Dr. Kirsten Haastert-Talini

Prof. Dr. Gerd Bicker

1st Evaluation: Prof. Dr. Wolfgang Baumgärtner

Department of Pathology, University of Veterinary Medicine

Hannover, Germany

Prof. Dr. Kirsten Haastert-Talini

Institute of Neuroanatomy, Medical School Hannover, Germany

Prof. Dr. Gerd Bicker

Institute of Animal ecology and cell biology, University of

Veterinary Medicine Hannover, Germany

2nd

Evaluation: Prof. A. Gröne

Department of Pathobiology, University of Utrecht, The

Netherlands

Date of final exam: 6th

November 2015

Muhammad Nadeem has received financial support from Higher education commission

Islamabad, Pakistan and DAAD Germany

To my family

Parts of the thesis have been published in peer-reviewed

journals previously:

AGUILAR-CALVO, P., FAST, C., TAUSCHER, K., ESPINOSA, J.C., GROSCHUP, M.H.,

NADEEM, M., GOLDMANN, W., LANGEVELD, J., BOSSERS, A., ANDREOLETTI, O.,

and TORRES, J.M. (2015):

Effect of Q211 and K222 PRNP Polymorphic Variants in the Susceptibility of Goats to Oral

Infection with Goat Bovine Spongiform Encephalopathy.

J Infect Dis. DOI: 10.1093/infdis/jiv112.

Parts of the thesis have been presented as poster at

congresses:

MUHAMMAD NADEEM, VERENA HAIST, CHRISTINE FAST, MARTIN H.

GROSCHUP, WOLFGANG BAUMGÄRTNER:

Ultrastructural pathology of the peripheral nervous system in caprine prion diseases. 2nd

International Workshop of Veterinary Neuroscience, Hannover, Germany, March 20-22,

2014

MUHAMMAD NADEEM, VERENA HAIST, CHRISTINE FAST, MARTIN H.

GROSCHUP, WOLFGANG BAUMGÄRTNER:

Ultrastructural pathology of the peripheral nervous system in caprine prion diseases. 2nd

N-

RENNT Symposium on Neuroinfectiology, Hannover, Germany, February 16-17, 2015.

Contents I

CHAPTER 1 INTRODUCTION OF TRANSMISSIBLE SPONGIFORM

ENCEPHALOPATHIES (TSEs) ............................................................................... 2

1.1. DEFINITION .............................................................................................................. 2

1.2. HISTORY .................................................................................................................... 2

1.3. CAUSATIVE AGENT ............................................................................................... 5

1.3.1. THE PRION THEORY ................................................................................ 5

1.3.2. NOMENCLATURE ...................................................................................... 6

1.3.3. PrPSc

FORMATION ...................................................................................... 7

1.3.4. PATHOGENESIS OF TSEs ......................................................................... 9

1.3.5. PATHOLOGICAL CHARACTERISTICS OF TSEs .............................. 11

1.4. CLINICAL MANIFESTATIONS............................................................................ 12

1.5. RODENT MODELS OF PRION DISEASES AND BRAIN PATHOLOGY IN

HAMSTERS ............................................................................................................. 12

1.5.1. CONVENTIONAL RODENT MODELS .................................................. 12

1.5.2. TRANSGENIC RODENTS MODELS ...................................................... 14

1.5.2 .1. PrP KNOCK-OUT MODELS ....................................................... 17

1.5.3. BRAIN PATHOLOGY IN HAMSTERS .................................................. 17

1.5.3 .1. STRAIN VARIATIONS ................................................................. 18

1.6. SYNAPTIC PATHOLOGY IN NEURODEGENERATIVE DISEASES ............ 20

1.6.1. MECHANISMS OF SYNAPTIC DYSFUNCTION ................................. 21

1.6.1.1. ROLE OF MITOCHONDRIA IN SYNAPTIC

DEGENERATION ......................................................................... 22

1.7. AIMS OF THE STUDY ............................................................................................ 24

CHAPTER 2 EFFECT of Q211 and K222 PRNP POLYMORPHIC VARIANTS IN THE

SUSCEPTIBILTY OF GOATS TO ORAL INFECTION WITH GOAT

BOVINE SPONGIFORM ENCEPHALOPATHY................................................. 25

CHAPTER 3 BSE INFECTION OF GOATS ALTERS NEUROFILAMENT

PHOSPHORYLATION STATUS OF SPINAL CORD AXONS ......................... 29

CHAPTER 4 DISCUSSION ............................................................................................................ 43

CHAPTER 5 SUMMARY ................................................................................................................ 51

CHAPTER 6 ZUSAMMENFASSUNG ........................................................................................... 55

CHAPTER 7 REFERENCES .......................................................................................................... 61

CHAPTER 8 ACKNOWLEDGEMENTS ...................................................................................... 89

II List of tables and figures

Table 1 TYPICAL FEATURES OF TSEs INCLUDING YEAR OF FIRST

DESCRIPTION, ROUTE OF TRANSMISSION, CLINICAL SIGNS AND

OTHER FEATURES .................................................................................................. 3

Table 2 NOMENCLATURE OF DIFFERENT TRANSMISSIBLE

SPONGIFORM ENCEPHALOPATHIES (TSES) ................................................. 6

Table 3 ORIGIN AND DIVERSITY OF EXPERIMENTAL TSE STRAINS

IN RODENTS ........................................................................................................... 13

Table 4 MOUSE MODELS TO STUDY PRION DISEASES MECHANISMS ............... 16

Fig. 1 FUNCTIONS OF NORMAL CELLULAR PRPC ................................................... 7

Fig. 2 CONVERSION OF NORMAL PRPC INTO ABNORMAL MISFOLDED

PRPSC

........................................................................................................................... 9

Abbreviation list III

Abbreviation list

TSEs = transmissible spongiform encephalopathies

BSE = bovine spongiform encephalopathy

CWD = chronic wasting disease

TME = transmissible mink encephalopathy

FSE = feline spongiform encephalopathy

CJD = Creutzfeldt-Jakob disease

sCJD = sporadic Creutzfeldt-Jakob disease

vCJD = variant Creutzfeldt-Jakob disease

Prion = proteinaceous infectious particle

PrPC

= normal cellular prion protein (C for cellular)

PrPSc

= abnormal prion protein (Sc for Scrapie)

Ov = ovine

Mk = mink

MDe = mule and deer

Bov = bovine

Fe = feline

Nya = nyala

Hu = human

PRNP = prion protein gene in human

GPI = glycosylphosphatidylinositol

CNS = central nervous system

PK = proteinase kinase

LRS = lymphoreticular system

SNS = sympathetic nervous system

ENS = enteric nervous system

GMCG = ganglion mesenterium craniale/ganglion coeliacum complex

DMNV = dorsal motor nucleus of the vagus

SHa = Syrian hamsters

SSBP = sheep scrapie brain pool

Tg = transgenic

WT = wild type

HY = hyper

DY = drowsy

SNAP25 = synaptosomal-associated protein of 25kDa

DPI = days post infection

PSD = post synaptic density

NMDA-R = N-methyl-D-aspartate-receptor

AMPA-R = α-amino-3-hydroxy-5-methyl-isoxazoleproprionic acid receptor

mGluR = metabotropic glutamate receptor

CSP = synaptophysin cysteine string protein

SNARE = soluble NSF attachment protein receptor

COX = cytochrome c oxidase

β-APP = beta amyloid precursor protein

CNPase = 2',3'-cyclic-nucleotide 3'-phosphodiesterase

Iba 1 = ionized calcium-binding adapter molecule

MAP 2 = microtubule associated protein

MHC Class II = major histocompatibility complex

P75NTR

= low affinity neurotrophin receptor p75

IV Abbreviation list

pNF = phosphorylated neurofilament

nNF = non-phosphorylated neurofilament

Tau 1 = tau protein

Chapter 1 1

Chapter 1

Introduction

2 Chapter 1

CHAPTER 1

1. Introduction of transmissible spongiform encephalopathies (TSEs)

1.1. Definition

Transmissible spongiform encephalopathies (TSEs) are also known as prion diseases. This is

a group of progressive conditions/syndromes that affects the nervous system of humans and

various animal species. They include bovine spongiform encephalopathy (BSE), scrapie,

chronic wasting disease (CWD), transmissible mink encephalopathy (TME), feline

spongiform encephalopathy (FSE), and others (CHESEBRO, 2003; LIBERSKI, 2012; LEE et

al., 2013). In humans, among others, Creutzfeldt-Jakob disease (CJD) with a sporadic (sCJD),

a variant (vCJD, the term new variant CJD is also used, here we will use the term vCJD), an

iatrogenic and a familial subtype exists (HAÏK and BRANDEL, 2014). The characteristic

features of TSEs comprise a long incubation period, multifocal spongiform changes,

astrogliosis, neuronal loss, and absence of inflammatory reaction in brain tissue

(CHESEBRO, 2003; IULINI et al., 2012).

The cause of TSEs is still under debate but it has been widely accepted that a transformed

host protein called prion (proteinaceous infectious particle), a novel type of infectious agent,

represents the etiology of these progressive diseases (PRUSINER, 1982).

1.2. History

As far as history about TSE diseases is concerned, scrapie was the first naturally acquired

prion disease known to the globe up to the 18th century. Scrapie transmission to healthy

sheep and goats from an affected sheep was reported during the 20th century (CUILLE and

CHELLE, 1936; CUILLE and CHELLE, 1939; CHELLE, 1942; DETWILER, 1992).

Transmissible mink encephalopathy (closely resembles L-type bovine spongiform

encephalopathy) as a food-borne disease of mink was first reported in the USA (MARSH and

HADLOW, 1992) and was later on detected in several other parts of the world

(SIGURDSON and MILLER, 2003). Kuru in humans was for the first time reported in 1957

and infection occurred most likely due to ritualistic cannibalism (ingestion of brain tissue) in

Papua New Guinea (GAJDUSEK and ZIGAS, 1957). The first case of feline spongiform

encephalopathy (FSE) was reported in a domestic cat in early 1990s (WYATT et al., 1990;

Chapter 1 3

BENCSIK et al., 2009), Chronic wasting disease (CWD) was firstly reported in 1967 as a

clinical syndrome of unknown etiopathogenesis in a restrained mule deer, originating from a

free-ranging population in Colorado, USA. A similar syndrome was identified in Wyoming

(USA) in 1978, and a spongiform encephalopathy was observed in the same year in affected

animals from several wildlife facilities in Colorado (WILLIAMS and YOUNG, 1980).

Bovine spongiform encephalopathy (BSE) was first reported in the UK in 1986 in cattle.

Recycled meat and bone meal from sheep with scrapie may be the cause for BSE in cattle.

Cattle being fed the remains of other cattle in form of meat and bone meal caused the spread

of the infectious agent (WELLS et al., 1987; WILESMITH et al., 1988). As a result a large

epidemic occurred that affected more than 182,000 cattle in 13 European countries, Canada,

the USA and Japan (GAVIER-WIDEN et al., 2005). Due to the implementation of rapid

screening tests and active surveillance (ANONYMOUS, 2001), BSE was also identified in

those European countries that were previously supposed to be BSE disease free countries.

Recently, outbreaks of TSE have been reported in small ruminants in a wide range of

countries, with the exceptions of Australia and New Zealand (GAVIER-WIDEN et al., 2005).

The typical features of TSEs in different animals have been shown in table 1.

Table 1. Typical features of TSEs including year of first description, route of

transmission, clinical signs and other features (adapted from RICHARD et

al., 2000 with modification)

TSEs in

Animals Year of first description

Route of

transmission

Typical clinic and

other features

References

Scrapie (sheep

and goats)

circa 1750 Acquired

(ingestion),

horizontal

transmission,

vertical

transmission

uncertain

Ataxia, pruritus Cuille and

Chelle, 1936;

Cuille and

Chelle, 1939;

Chelle, 1942;

Konold et al.,

2010

Sporadic

Creutzfeldt–

Jakob disease

(CJD)

1920 Unknown.

Somatic

mutation or

spontaneous

conversion of

PrPc into PrP

Sc

Dementia, myoclonus,

variable ataxia;

spongiform changes;

variable amyloid plaques

(about 15% of cases)

Kretzschmar,

1993, Richard

et al., 2000

4 Chapter 1

Continuation of table 1

TSEs in

Animals Year of first description

Route of

transmission

Typical clinic

and other

features

References

Familial

Creutzfeldt–

Jakob disease

1924 Associated with

germline

mutations in

PRNP gene with

autosomal-

dominant pattern

of expression

Same as CJD,

longer survival;

amyloid plaques

more common

Gambetti et al.,

2004 ; Richard

et al., 2000

Gerstmann-

Straussler-

Scheinkes

syndrome

1936 Associated with

germline

mutations in

PRNP gene,

autosomal-

dominant pattern

Familial, ataxia,

dementia;

amyloid plaques

Ravilochan and

Tyler, 1992

Kuru 1957 Ritualistic

cannibalism,

ingestion of brain

tissue, skin

Ataxia, tremor,

cranial nerve

abnormalities;

amyloid plaques

are common

Gajdusek and

Zigas, 1957

Transmissible

mink

encephalopathy

(mink)

1965 Acquired

(ingestion) but

source unknown

sheep with

scrapie

Ataxia,

somnolence,

seizures

Richard et al.,

2000

Chronic wasting

disease (deer,

elk)

1980 Acquired

(ingestion),

horizontal

transmission,

vertical

transmission

unclear

Altered behavior,

excessive

salivation,

wasting;

florid amyloid

plaques

Mathiason et al.,

2006

Fatal familial

insomnia

1986 Associated with

germline

mutations in

PRNP gene/

mutation at

PRNP,

autosomal-

dominant

inheritance,

Insomnia,

dysautonomia,

ataxia,

myoclonus, late

mild dementia;

minimal

vacuolation, no

plaques,

PrPSc

difficult to

detect

Ravilochan and

Tyler, 1992

Bovine

spongiform

encephalopathy

(cattle, zoo

ruminants)

1987 Ingestion of BSE-

contaminated

meat and bone

meal

Spontaneous

diseased cow

Ataxia, wasting Richard et al.,

2000, Wilesmith

et al., 1988

Chapter 1 5

Continuation of table 1

TSEs in

Animals Year of first description

Route of

transmission

Typical clinic and

other features

References

Feline

spongiform

encephalopathy

1990 Ingestion of

BSE-

contaminated

food

Altered behavior, ataxia Wyatt et al.,

1990; Bencsik

et al., 2009

Variant

Creutzfeldt–

Jakob

disease (vCJD)

1996 Ingestion of

BSE-

contaminated

food. transfusion

of blood from

vCJD-infected

blood donor.

Young age at onset;

psychiatric

presentation,

dysesthesias, ataxia;

florid amyloid plaques

Will et al.,

1996

Sporadic

familial

insomnia

1999 Same as fatal

familial

insomnia but

negative family

history; no

mutation

identified in

either PRNP

gene

Same as fatal familial

insomnia Mastrianni et

al., 1999

BSE= bovine spongiform encephalopathy, CJD = Creutzfeldt-Jakob disease, Sc = scrapie

1.3. Causative agent

1.3.1. The prion theory

The causative agent of transmissible spongiform encephalopathies is still not completely

understood but several theories have been proposed. Historically researchers assumed the

causative agent to be a slow virus infection but no virus was isolated from brain tissues of

TSE affected animals (ALPER et al., 1966; PRUSINER, 1982; RIESNER, 2003).

Furthermore the treatments that caused inactivation of most viruses and nucleic acid (e.g.

heat, ionization radiation, alcohol, formalin, some proteases and nucleases) remained

ineffective in controlling the infectious nature of the TSE agent (RICHARD et al., 2000). A

protein as a causative agent for scrapie was firstly proposed in the mid-1960s (PATTISON

and JONES, 1967; ALPER et al., 1966). GRIFFITH (1967) tried to explain the replication

mechanism of proteins devoid of nucleic acids. Further studies confirmed that the chemical

properties of the infectious agent are similar to protein molecules, thus supporting the

protein-only hypothesis (PRUSINER et al., 1980). In 1982, PRUSINER demonstrated a small

proteinaceous infectious (prion) particle as the primary component of the scrapie agent and

6 Chapter 1

introduced the term prion. Prions consist of a misfolded prion protein (PrP) designated as

PrPSc

(Sc for scrapie); whereas the normal cellular prion protein is designated as PrPC

(C for

cellular prion protein). This protein-only model of the prion theory suggests that a molecular

mechanism is involved in replication of the TSE agent by which abnormally folded PrPSc

serves as a template or catalyst which recruits cellular PrP and transforms it to its 3-

dimensional infectious structure (BEEKES and McBRIDE, 2007; MORINET, 2014).

1.3.2. Nomenclature

The nomenclature of PrP species is complicated. The normal cellular isoform is designated as

PrPC whereas PrP

Sc stands for the proteinase K resistant, misfolded protein that remains

insoluble in denaturing detergent. However, there are exceptions in few diseases occasionally

the pathological isoform of PrP fails to show proteinase K resistance (GABIZON et al.,

1996). Recently, the term PrPTSE

has been introduced for diseases associated PrP from TSE

infected individuals to avoid the confusion with the complex PrP nomenclatures e.g. PrPCJD

,

PrPCWD

, PrPSEN

, PrPres

, PrPSc

and PrPBSE

. The currently used nomenclature for the different

prion isoforms is shown in table 2 (BROWN and CERVENAKOVA, 2005).

Table 2. Nomenclature of different transmissible spongiform encephalopathies (TSEs)

Disease name Natural host Prion name PrP isoform

Non-human mammals

Scrapie Sheep and goat Scrapie prion OvPrPSc

Transmissible mink

encephalopathy (TME) Mink TME prion MkPrP

Sc

Chronic wasting disease

(CWD)

Elk, White-tailed deer,

Mule Deer and Red Deer CWD prion MDePrP

Sc

Bovine spongiform

encephalopathy (BSE) Cattle BSE prion BovPrP

Sc

Feline spongiform

encephalopathy (FSE) Cat FSE prion FePrP

Sc

Exotic ungulate

encephalopathy (EUE) Nyala and Greater kudu EUE prion NyaPrP

Sc

Human diseases

Kuru

Humans

Kuru prion

HuPrPSc

Creutzfeldt-Jakob disease

(CJD) CJD prion

(New) Variant Creutzfeldt-

Jakob disease (vCJD,

nvCJD)

vCJD prion

Gerstmann-Sträussler-

Scheinker syndrome (GSS) GSS prion

Chapter 1 7

Ov = ovine, Mk = mink, MDe = mule and deer, Bov = bovine, Fe = feline, Nya = Nyala, Hu

= human, Sc = scrapie

1.3.3. PrPSc

formation

The normal cellular prion protein (PrPC) is a glycoprotein primarily present on the

membranes of neurons, glial cells and in various organs including uterus, placenta, thymus,

heart, lung, muscle and gastrointestinal tract (BUDKA, 2003). PrPC is encoded by the prion

protein gene (PRNP) and is highly conserved among different species (van RHEEDE et al.,

2003). PrPC is attached to the cell surface by using a glycosylphosphatidylinositol (GPI)

anchor (RIESNER, 2003). The normal cellular prion is an α-helical conformational copper-

binding protein with an approximately 220 amino acid residue (RIEK et al., 1996;

HORNEMANN et al., 1997; GAVIER-WIDEN et al., 2005). In the central nervous system

(CNS) PrPC

has several functions (Fig.1) including neuronal survival, neurite outgrowth,

synapse formation, maintenance of myelinated fibers and protection against apoptosis or

oxidative stress (WESTERGARD et al., 2007; AGUZZI et al., 2008).

Fig. 1. Functions of normal cellular PrPC (adapted with modifications from AGUZZI et al.,

2008 and learn.genetics.utah.edu)

The abnormal, comparatively protease-resistant, ß-pleated sheet-rich isoform of PrPC is

traditionally called PrP scrapie (GAVIER-WIDEN et al., 2005). The formation of the

pathological isoform of prion protein is thought to be activated either by a mutation of the

normal cellular prion protein gene or by the transmission of pathogenic prion isoforms

(DeMARCO and DAGGETT, 2004; AGUZZI et al., 2008).

8 Chapter 1

The posttranslational modification of PrPC into the abnormal pathological form occurs

through a process of conformational changes, whose mechanism remains elusive until now.

Studies using transgenic mouse models have shed some light on genetic and biochemical

mechanisms responsible for the conversion. According to these models, PrPC

is converted

into PrPSc

by the formation of a PrPC/PrP

Sc complex, but this complex has never been isolated

in pure form. Therefore, it remains unclear whether PrPC binds to one or more additional

macromolecules during the conversion process (PRUSINER et al., 1990; MEIER et al., 2003;

AGUZZI et al., 2008). During conformational changes, ß-pleated sheets become dominant

over the α-helical structure, resulting in a characteristic fibrillar aggregated structure in the

brain as seen in many TSE diseases (PRUSINER, 1998; FRASER, 2002; PRUSINER, 2013).

Spectroscopic measurements of PrPC from healthy hamster brains demonstrated that PrP

C is

mainly composed of α-helices (42%) with negligible amount of β-sheets (3%). On the other

hand, PrPSc

recovered from scrapie infected hamster brain consists of 43% β-sheets and 30%

α-helices (PAN, 1993).

PrPC is normally present on the neuronal cell surface in contrast; PrP

Sc is found in the

cytoplasm of affected cells and shows high resistance against common sterilization methods

(e.g. autoclaving, heat and radiation), proteolytic enzymes, and conventional desinfectants

including alcohol, formalin, and phenol (BELLINGER et al, 1987; BELL and IRONSIDE,

1993).

Once the abnormal isoform is formed or acquired (Fig. 2), it catalyzes the conversion of PrPC

molecules into PrPSc

through an autocatalytic process (CAUGHEY and RAYMON, 1991).

Breakage of these provides more PrPSc

templates for further conversion of the cellular prion

protein in neighbouring neuronal cells. Thus proteinase kinase (PK)-resistant, non-degradable

PrPSc

aggregates in the neuronal tissues are formed and serve as the most effective marker of

prion diseases (McKINLEY et al., 1983).

Chapter 1 9

Fig. 2. Conversion of normal PrPC into abnormal misfolded PrP

Sc (adapted from AGUZZI

and POLYMENIDOU, 2004 with modification).

(1) The refolding or template assistance model highlights the interaction between an

externally introduced misfolded protein form (PrPSc

) and normal cellular prion protein (PrPC).

PrPSc

utilizes the normal cellular prion protein as template to transfer itself into further PrPSc

.

A high energy barrier may avoid further spontaneous conversion of the misfolded form. (2)

The seeding or nucleation-polymerization model proposes that the normal cellular prion

protein and the abnormal misfolded form are in a thermodynamic equilibrium. When several

monomeric PrPSc

molecules are mounted into a highly ordered and infectious seed, they can

recruit further PrPSc

and finally aggregate in the form of amyloid.

1.3.4. Pathogenesis of TSEs

The pathogenesis of prion diseases is also poorly understood. Among naturally acquired

prion diseases, scrapie and BSE are the most relevant forms because of their economic

importance. Futhermore, BSE plays an important role due to its transmission to humans and

by causing vCJD (SCOTT et al., 1999; BEEKES and McBride, 2007). Infection begins upon

ingestion of the TSE agent and subsequent invasion of the gut by the pathogen in the majority

of cases of scrapie, BSE, TME, CWD, vCJD and kuru (GAJDUSEK, 1977). Ritualistic

cannibalism seems to be one route of kuru transmission, and BSE contaminated foodstuff

ingestion served as a source of vCJD. In comparison to other TSEs, scrapie and CWD are the

only TSEs that are not only transmissible but also contagious. Infected placenta, abraded skin

Heterodimer Homodimer

Amyloid; not

essential for

replication

Template assisted refolding

Conversion

prevented

by energy

barrier

Equilibrium

between

both forms

PrPC

PrPSc

PrPC

PrPSc

Seed formation

(very slow) Recruitment of

PrPSc

(fast) Infectious

seed

Amyloid Fragmentation

into seeds

Seeding Nucleation

1

2

10 Chapter 1

and flesh of dead animals (in the form of meat and bone meal) are considered the major cause

of horizontal or vertical transmission of scrapie in sheep (BROWN and GAJDUSEK, 1991;

RACE et al., 1998; DETWILER and BAYLIS, 2003). Additionally mites, fly larvae and

pupae serve as living harbours of ingestible infectivity (WISNIEWSKI et al., 1996; POST et

al., 1999). Recently, prion agents were found in CWD infected cervid saliva (MATHIASON

et al., 2006). Along ingestion, scarification of skin or gums is also an important route of agent

entry into the body. In the case of kuru, transdermal/conjuctival invasion served as an

alternative natural source of infection (GOODFIELD, 1997). In early stages of disease

progression, infectious prion agents cross the mucous membrane barriers and can be detected

in tonsils, Peyer’s patches, and lymph nodes of the alimentary canal (ANDRÉOLOTTI et al.,

2000; SPRAKER et al., 2002; JEFFREY and GONZÁLEZ, 2004; WELLS et al., 2005). This

phenomenon of early lymphoid invasion has been demonstrated experimentally as early as 6

weeks after infection in CWD (SIGURDSON et al., 1999) and at 3 months of age in naturally

occurring scrapie in lambs (ANDRÉOLOTTI, 2004).

After invasion of the infectious agent, a replication period lasting from months to years takes

place in the lymphoreticular system (LRS) involving spleen and lymph nodes in most cases

of TSEs. However, in BSE and in some scrapie cases, there is little involvement of the LRS

(JEFFREY and GONZÁLEZ, 2004; WELLS et al., 2005). After incubation the infectious

prion agent spreads towards the brain, where it progressively aggregates, resulting in fatal

neurodegenerative alterations. The mechanisms involving the spread of prions from the

alimentary tract or tonsils to the brain are inadequately investigated. Hematogenous and

retrograde axonal routes, involving fibers innervating lymphoid tissues or the autonomic

nervous fibers of the digestive tract, have been implicated (SIGURDSON et al., 1999;

ANDRÉOLOTTI et al., 2000) resulting in a model of neuroimmune invasion that comprises

two phases. The first phase is characterized by the widespread colonization of

lymphoreticular organs by a mechanism that depends on B lymphocytes and follicular

dendritic cells. The second phase involves the expression of PrPSc

in the peripheral

sympathetic nervous system (SNS) nerves and results in the prion distribution in the CNS.

The neuronal spread of prion infectious agents from the enteric and peripheral nervous

system to the spinal cord after oral uptake of the TSE agent from the gut was first proposed

after an intra-gastric scrapie challenge to mice (KIMBERLIN and WALKER, 1989). Later,

hamster adapted 263K scrapie served as a model to observe the neuronal spread of the prion

Chapter 1 11

agent from the alimentary canal to the brain after oral uptake (BEEKES et al., 1998;

McBRIDE et al., 2001). It was shown that the N. splanchnicus and N. vagus of sympathetic

and parasympathetic systems; respectively, are the main routes of prion spread from the gut

to the CNS. Efferent and afferent nerve fibers are used to reach either to the thoracic spinal

cord (splanchnic nerves) or the solitary tract nucleus and the dorsal motor nucleus of the N.

vagus. Centripetal and centrifugal spread of the prion agent to the cervical and lumbal spinal

cord originating from the thoracic spinal cord (McBRIDE et al., 2001; BALKEMA-

BUSCHMANN et al., 2011; KAATZ et al., 2012; McGOVERN et al., 2015). Sheep and

goats naturally or experimentally infected with prion disease have shown a significant

propagation of the scrapie agent in lymphoid organs including Peyer´s patches, spleen and

lymphoid ganglions during the early stage of infection. With the progression of the disease

the agent is present in several tissue and fluids with high infectious titers in the brain. In the

case of cattle TSE, infectivity is mainly detected in different parts of the CNS, the peripheral

nervous system and autonomic nervous system.

1.3.5. Pathological characteristics of TSEs

Neuropathologically, TSEs are characterized by spongiosis or vacuolation in the neuropil

(vacuolation of neuronal processes), and/or neuronal bodies showing single or multiple

vacuoles in the perikarya of neurons (WELLS et al., 1987; WELLS et al., 1989; WILLIAMS

and YOUNG, 1993; SPRAKER et al., 2002). PrPSc

aggregation and accumulation in neurons

and glial cells in the brain is the characteristic feature of TSE and can be detected earlier than

vacuolar changes (JEFFREY and GONZÁLEZ, 2004; SPRAKER et al., 2004). Other

remarkable changes including neuronal cell death or loss, astrocyte proliferation and amyloid

plaque formation are variably seen in some forms of human and animal TSEs (BUDKA et al.,

1995; WELLS and WILESMITH, 1995; LIBERSKI et al., 1998; LIBERSKI and BUDKA,

1999; FRASER, 2002). Classical inflammatory responses against the infectious isoform of

PRPTSE

are not initated presumable since the faulty protein is not recognized as foreign

material by the immune system (GAVIER-WIDEN et al., 2005). The mechanism(s) causing

brain damage through accumulation of PrPSc

have not been fully elucidated. Although a

noticeable accumulation of the pathological isoform is also present in the lymphoid tissue, no

histological alterations in lymphoid tissues have been observed (GAVIER-WIDEN et al.,

2005).

12 Chapter 1

1. 4. Clinical Manifestations

Infected animals may develop signs of the disease slowly and many months and years after

primary exposure. In cattle it may take 2 to 8 years from the time an animal becomes infected

until it shows first signs of the disease. Signs include a change in attitude and behaviour,

gradual uncoordinated movements, trouble in standing and walking, weight loss despite

normal appetite, and decreased milk production. From the onset of signs, the animal

deteriorates until it either dies or is destroyed. This disease process may take from 2 weeks to

6 months after first initial clinical signs have been noticed. Similar symptoms consisting of

muscle spasms, lack of muscle control, deteriorating problems with memory may develop in

humans.

1.5. Rodent models of prion disease and brain pathology in hamsters

Due to the unavailability of cell culture systems for pathogenetic studies of prion diseases,

conventional or transgenic animal models provide an opportunity to study most aspects of

prion propagation and infectivity (WATTS and PRUSINER, 2014). Previous in vivo studies

on TSEs were carried out mostly in the natural host species. Rodent models expressing

cellular prion proteins from different species provide the opportunity to study the disease in a

more formalized manner. These models help to understand the neuropathological

mechanisms on the molecular level, normal functions of PrPC, species barrier mechanisms,

cell specificity, role of glycosylation, prion agent spread mechanism and interaction between

PrPC and PrP

Sc (GROSCHUP and BUSCHMANN, 2008). They also shed some light into

mapping of prion protein segments which are involved in prion conversion and replication

and helped to understand the role of the host prion gene in the genetic control of the disease

(BARON, 2002; GROSCHUP and BUSCHMANN, 2008).

1.5.1. Conventional rodent models

Since the first transmission of scrapie to mice, the use of animal models has laid the basis for

a more comprehensive understanding of prion diseases (CHANDLER, 1961). Over the years,

rats, golden hamsters and voles were also used as animal models (CHANDLER, 1971;

CHANDLER and FISHER, 1963; CHANDLER and TURFREY, 1972). These animal

species provide a great opportunity to study disease characteristics in more detail due to the

Chapter 1 13

short incubation time compared to the natural hosts (see table 3 for brief description of these

models and their origin). Initial experiments performed on Syrian hamsters (SHa) allowed the

prediction of the prion protein theory (PRUSINER et al., 1980). Successful experimental

transmission of prion isolates has also been reported to non-human primates including

monkeys (macaque and squirrel), lemurs and chimpanzees (GIBBS et al., 1994; WILLIAMS

et al., 2007; LASMÉZAS et al., 2005).

Table 3. Origin and diversity of experimental TSE strains in rodents (adapted from

BERINGUE et al., 2008).

Name/abbreviation

of strains

Origin Passage in

intermediate

species

Host References

ME7

(Prn-a)

Sheep scrapiea None Mouse Bruce, 1993; Bruce

et al., 2002

87A Sheep scrapiea None Mouse (Prn-

a)

Bruce and

Dickinson, 1987

221C Sheep scrapiea None Mouse (Prn-

a)

Bruce et al., 2002

87V Sheep scrapiea

SSBP/1b

None, goatsc Mouse (Prn-

b)

Bruce, 1993; Bruce

et al., 2002

79A SSBP/1b goats

c Mouse (Prn-

a)

Bruce, 1993

79V SSBP/1b goats

c Mouse (Prn-

b)

Bruce, 1993

139Ad SSBP/1b goats

c Mouse (Prn-

a)

Chandler and

Fisher, 1963

14 Chapter 1

Continuation of table 3

Name/abbreviation

of strains

Origin Passage in

intermediate

species

Host References

22C SSBP/1b None, goats

e Mouse (Prn-a) Bruce, 1993

22H Uncloned 22C None, goatse Mouse (Prn-b) Bruce, 1993

22L SSBP/1b None Mouse (Prn-a) Bruce, 1993

22A SSBP/1b None Mouse (Prn-b) Bruce, 1993

22F Cloned 22A None Mouse (Prn-a) Bruce, 1993

301Cf BSE Direct or not Mouse (Prn-a) Bruce et al.,

1994

301V BSE Direct or not Mouse (Prn-b) Bruce et al.,

1994

139H Cloned 139A None Syrian hamster Kimberlin et

al., 1987

263Kg SSBP/1b goats

c, mice,

rats

Syrian hamster Kimberlin and

Walker , 1977

ME7-H Cloned ME7 None Syrian hamster Kimberlin and

Walker, 1989

HYh TMEi None Syrian hamster Bessen and

Marsh, 1992

DY TMEi None Syrian hamster Bessen and

Marsh, 1992

a = Field isolate; b = SSBP/1: sheep scrapie brain pool 1; c = Passage of SSBP/1 through

goats: ‘drowsy’ goat source; d = also termed Chandler; e = Passage of SSBP/1 through goats:

‘scratching’ goat source; f = not known if 301C yields to 301V in Prn-b mice; g = Sc237 is a

subclone of 263K; h = similar to 263K?; i = TME: Stetsonville isolate, BSE: bovine

spongiform encephalopathy

1.5.2. Transgenic rodent models

Mouse transgenesis has made enormous contribution to prion research (table 4). SCOTT et

al. (1989) generated transgenic mice overexpressing hamster PrPC that proved to be

susceptible to 263K hamster prions in contrast to conventional mice. Nowadays a series of

transgenic mouse lines is available which express mink, human, caprine, bovine, ovine,

Chapter 1 15

cervid, and mouse PrPC (table 4). In addition, SHMERLING et al. (1998), generated mouse

models expressing transgenic PrPC with amino-proximal deletions at residues 32-121 or 32-

134. These mice showed severe ataxia along with neuronal death in the granular layer of the

cerebellum as early as 1-3 months after birth. This deficiency was recovered by introducing

one copy of a wild-type PrP gene. PrPC transgenic mice with deleted individual regions of the

putative secondary structure demonstrated that lacking of one of the C-terminal helices lead

to the incidence of CNS failures. This accumulation of PrP within neurons as cytoplasmic

inclusions (MURAMOTO et al., 1997) highlights the probable role of α-helix in protein

stability and normal trafficking. Glycosylphosphatidylinositol lacking transgenic (GPI–Tg)

mice, inoculated with scrapie prion exhibited susceptibility to infection but an altered clinical

disease manifestation and PrPSc

deposits were noticed (CHESEBRO et al., 2005). On the

other hand, the wild type mice inoculated with the scrapie prion generated the normal profile

of prion disease; thus, highlighting the possible role of the GPI anchors in disease outcome.

Transgenic mice expressing PRNP with point mutations, insertions, or deletions exhibited

phenotypically a similar spongiform diseases (SIGURDSON et al., 2009). It has been

observed that a moderate overexpression in transgenic mPrP (170N, 174T) mice (a mouse

PrP with two point mutations that affect the structure of its globular domain) resulted in the

generation of spongiform encephalopathy with cerebral PrPSc

plaques. This genetic disease

was restored by intracerebrally inoculation of brain homogenate to tga20 mice

overexpressing wild type (WT-PrP) PrP (SIGURDSON et al., 2009). Transgenic mice allow

to study the pathogenesis of several mutations related to different forms of genetic TSEs, the

transmission barrier phenomenon (AGUILAR-CALVO et al., 2014) and hence to assess the

relative risk of each TSE strain for humans. For example, tg650 mice expressing human PrP

Met129 were inoculated with field isolates of different forms of BSE. Unlike the classical

BSE agent, L-type BSE emerged to proliferate in these mice with no obvious transmission

barrier, whereas H-type prions were unable to infect these mice (BERINGUE et al., 2008).

16 Chapter 1

Table 4. Mouse models to study prion diseases mechanisms (adapted from BARON, 2002)

BSE: bovine spongiform encephalopathy, Vcjd: variant Creutzfeldt-Jakob disease

Mouse models Field of application References

Wild-type mice

Transmission and species barrier

Fraser et al., 1992 ;

Chandler, 1961

Resistance to prion diseases

Race and Chesebro,

1998

Genetic control of prion diseases

Carlson et al., 1988 ;

Lloyd et al., 2001

Biological typing

Bruce, 1996; Fraser and

Dickinson 1973

Molecular typing

Baron and Biacabe,

2001

Putative role of associated factors in

strain formation Somerville et al., 2002

Knockout PrP 0/0

mice

Transmission studies and species barrier Prusiner et al., 1993

Studies of doppel (Dpl) protein Tuzi et al., 2002

Hamster transgenic

mice Transmission studies and species barrier Scott et al., 1989

Mouse transgenic

mice

Models of human genetic disease Manson et al., 1999

Transmission studies and species barrier Fischer et al., 1996;

Buschmann et al., 2000

Human transgenic

mice

Transmission studies of CJD Telling et al., 1994

Putative role of associated factors in

transmissibility Telling et al., 1995

Biological typing of Vcjd Hill et al., 1997

Molecular origin of strains Telling et al., 1996

Ovine transgenic

mice

Transmission of scrapie Crozet et al., 2001b

Biological typing of BSE and scrapie Crozet et al., 2001a

Bovine transgenic

mice

Transmission of BSE Buschmann et al., 2000

Chapter 1 17

1.5.2.1. PrP knock-out models

After development of the protein-only hypothesis, the generation of transgenic mouse strains

lacking PrPC

expression (PrP knockout mice) helped to understand the physiological function

of PrPC

and its role in neurodegenerative diseases in more detail. At least four lines of mice

lacking PrPC have been developed (WEISSMANN and FLECHSIG, 2003). Ablation of PrP

C

in these models did not result in major anatomical and developmental deficits; however, these

animals were resistant to scrapie challenge (BUELER et al., 1992; BUELER et al., 1993;

PRUSINER et al., 1993; SAILER et al., 1994; WEISSMANN and FLECHSIG, 2003). After

reintroduction of PrP transgenes in PrP knockout animals, the susceptibility to infection was

restored confirming a correlation between host PrPC and expression of TSE. Introduction of

multiple prion gene copies to the mouse genome leading to the overexpression of PrPC,

showed that the PrPC expression concentration plays an important factor for prion disease

susceptibility (PRUSINER et al., 1990; FISCHER et al., 1996; WEISSMANN and

FLECHSIG, 2003; UCHIYAMA et al., 2014).

1.5.3. Brain pathology in hamsters

Transmission of scrapie prions to golden hamsters was first reported by ZLOTNIK and

RENNIE (1965) using the ME7 strain of the scrapie agent. This was confirmed by

CHANDLER and TURFREY (1972), who successfully transmitted scrapie to Chinese

hamsters. Among the transmissible spongiform encephalopathies, the hamster 263K scrapie

prion model is a rapid and well characterized model (BOLTON et al., 1991). A low

concentration of prions can easily be detected with hamster prion models as compared to

other models which are less sensitive (BOLTON, 1998). Incubation periods of scrapie in

hamsters are remarkably short as compared to mouse models (KIMBERLIN and WALKER,

1977). Hamsters inoculated intracerebrally with a high dose of scrapie prions develop clinical

signs consisting of neurological dysfunction after 60-65 days. This is half of the incubation

time found in mice (BARINGER et al., 1983).

The prion burden remains stable in various CNS regions throughout the disease course and

precedes pathological changes. Histologically, the cerebrum shows minimal vacuolation in

the absence of astrogliosis prior to clinical signs. After the onset of clinical signs, severe

vacuolation with moderate astrogliosis has been observed in the cerebral cortex. Cerebellum,

18 Chapter 1

brain stem, and spinal cord display a moderate degree of vacuolation along with astrogliosis

(BARINGER et al., 1983).

1.5.3.1. Strain variations

Concerning the causative agents of TSEs some reservations remain. A foremost question is

whether the causative agents are exclusively composed of one specific abnormal isoform of

the normal cellular prion protein. A major problem for the protein-only hypothesis of prion

diseases has been how to explain the presence of multiple isolates or strains of prions. The

existence of different prion strains was first observed in goats after inoculation with sheep

brain homogenates (SSBP/1), which resulted in two different clinical disease phenotypes: a

scratching and a drowsy syndrome (PATTISON et al., 1959). Now several discrete strains of

naturally occurring sheep scrapie have been isolated in mice. Such strains are distinguished

by their biological properties including distinct incubation periods and lesion profiles in

defined inbred mouse lines (BRUCE et al., 1992). For instance, they can be serially

propagated in inbred mice with the same Prnp genotype. Moreover, strains can be re-isolated

in mice after passage in intermediate species with dissimilar PrP primary structures (BRUCE

et al., 1994). Usually, distinct strains of conventional pathogens including bacteria and

viruses are described by their difference in their nucleic acid genome. In the absence of such

a scrapie genome, alternate prospects must be considered. WEISSMANN`s (1991) “unified

hypothesis” suggested that strain characteristics could be encoded by a small cellular nucleic

acid, or “coprion.” According to this hypothesis the strain characteristics would be sensitive

to ultraviolet irradiation, but he failed to present such results. On the other hand, the protein-

only hypothesis proposed by GRIFFITH 1967 would have to explain how a single

polypeptide chain might encode multiple disease phenotypes. Evidently, understanding how a

protein-only infectious agent could convert such phenotypic information is of significant

biological importance (COLLINGE, 2001).

Strain specificity encoded by PrP itself was supported by the study of two distinct strains of

TME prions propagated in hamsters, designated as hyper (HY) and drowsy (DY). These

strains can be differentiated by differing biochemical properties produced by the accumulated

PrPSc

in the brains of affected hamsters (BESSEN and MARSH, 1992; COLLINGE, 2001).

With limited proteolysis, strain-specific migration patterns of PrPSc

were seen on

polyacrylamide gels. They were linked to different N-terminal ends of HY and DY PrPSc

Chapter 1 19

following protease treatment and involved differing conformations of HY and DY PrPSc

(BESSEN and MARSH 1994; COLLINGE, 2001). Several human PrPTSE

confirmations

related to different phenotypes of CJD have been identified (TELLING et al., 1996;

COLLINGE et al, 1996; COLLINGE, 2001). The different fragment sizes after proteinase K

treatment as seen on Western blots highlight the presence of different PrPSc

conformations

(SAFAR et al., 1998). Biochemically modified PrP served as candidates for the molecular

substrate of prion strain diversity. This aspect has been elaborated on studies with CJD

isolates. PrPTSE

fragment sizes and PrP glycoforms ratios (diglycosylated, monoglycosylated,

and unglycosylated PrP) were maintained in human PrP expression in transgenic mice

following passages. Additionally, transmission of human and bovine prions to wild type mice

results in murine PrPSc

with fragment sizes and glycoforms ratios corresponding to the

original inoculum (COLLINGE et al 1996; COLLINGE, 2001). Vcjd is distinct from

classical CJD on the basis of PrPSc

glycoforms ratios. Related ratios are also observed in BSE

in cattle and BSE transmitted to numerous other species. These observations intensely

support the protein-only hypothesis of infectivity and indicate that strain variation is

determined by the arrangement of PrP conformation and glycosylation. Moreover,

polymorphisms shown by the PrP sequence can affect the generation of specific PrPSc

conformers. As glycosylation happens before changing to PrPSc

, the diverse glycoforms ratios

may signify selection of specific PrPC glycoforms by PrP

Sc of diverse conformations.

Following such a hypothesis, PrP conformation would be the major factor determining the

strain type, with glycosylation as a secondary process. However, as it is observed that

different cell types glycosylate proteins differently, PrPSc

glycosylation forms might offer a

substrate for the neuropathological targeting that discriminates diverse prion strains

(COLLINGE et al 1996; COLLINGE., 2001). Specific PrPSc

glycoforms might replicate well

in neuronal populations expressing a similar PrP glycoforms on the cell surface. Such

targeting could also help to explain the different incubation periods that also allows

distinguishing strains. Subsequently, targeting of brain regions with higher levels of PrP

expression will likely yield shorter incubation periods (SAFAR et al., 1998). The results also

suggested that different conformations of PrPSc

could encipher properties of different prion

strains characterized by altered pathological behaviours (SAFAR et al., 1998). Furthermore,

it was shown that from a single source of a scrapie agent a mixture of strains could be

isolated (KIMBERLIN and WALKER, 1978). However, it is still unclear if these sub-strains

are stable in themselves or are dependent on the co-existence of their “partner strains”.

20 Chapter 1

1.6. Synaptic pathology in neurodegenerative diseases

Despite the significant importance of neuronal death in neuropathology of prion diseases, the

events and mechanism(s) that lead to neuronal dysfunction and ultimately neurodegeneration

remain inadequately understood. The suggestions of a possible correlation between cognitive

decline and synaptic loss in Alzheimer’s disease have opened new avenues in the prion field

too (TERRY et al., 1991). There is a growing body of evidence that indicates that synaptic

dysfunction plays an early and important role in the development and progression of prion

diseases and it may be an early key event in many neurodegenerative diseases (CLINTON et

al., 1993; CUNNINGHAM et al., 2003; JEFFREY et al., 2000; SISKOVA et al., 2009; REIS

et al., 2015).

Immunohistochemical, ultrastructural and cellular studies have demonstrated that pre-

synaptic terminals in brain synapses are enriched in PrPC

(FOURNIER et al., 1995;

HAEBERLE et al., 2000; BROWN, 2001). Early events in the development of prion diseases

involve synaptic loss associated with deposition of abnormal PrPSc

in synaptic boutons

especially in pre-synaptic terminals (JEFFREY et al., 2000). Neurotransmission and exosome

associated synaptic vesicle proteins e.g. synaptophysin and synapsin-I, and proteins of pre-

synaptic plasma membrane e.g. synaptosomal-associated protein of 25kDa (SNAP-25) and

syntaxin-I, are reduced in patients suffering from prion diseases (FERRER, 2002; FERRER

et al., 1999). These proteins play a vital role in exocytosis and neurotransmission, and some

of these proteins contribute for normal synaptic function. Therefore, it may be assumed that

pre-synaptic modulation is damaged in prion diseases (FERRER, 2002). However, reduction

in synaptic protein expression should not be considered as an exclusive cause of synaptic

loss. Impaired or abnormal protein synthesis or turnover may also represent a complementary

event in synaptic dysfunction (FERRER, 2002). Synaptic dysfunction is an essential and

constant feature of prion disease, irrespective of the existence or lack of spongiform changes,

neuronal loss and severe gliosis (CLINTON et al., 1993). In rodent models of

neurodegenerative diseases it is well documented that synaptic pathology precedes the

degeneration of neuronal cell bodies in the hippocampus (CUNNINGHAM et al., 2003;

JEFFREY et al., 2000; SISKOVA et al., 2009). Malformed electrophysiological recordings in

scrapie infected hamster hippocampal and cortical slices further substantiate the synaptic

alterations (BARROW et al 1999). The murine ME7 scrapie model was among the first to

present observations that allowed to distinct between synaptic dysfunction and neuronal cell

Chapter 1 21

death (JEFFREY et al., 2000; CUNNINGHAM et al., 2003). In this model, synaptic

degeneration within the stratum radiatum of the hippocampus is characterized by the

degeneration of the pre-synaptic terminal, proceeding to the loss or degeneration of the post-

synaptic dendritic spine. In addition, these changes occur in the absence of detectable

neuronal cell death (CUNNINGHAM et al., 2003; SISKOVA et al., 2009). Electron

microscopic studies in the murine model showed that synaptic dysfunction and loss

associated with PrPSc

preceded neuronal loss and clinical onset of disease. Scrapie infected

murine hippocampus revealed degenerated axon terminals at about 98 days post infection

(dpi), whereas definite clinical scrapie is apparent not before 226 dpi (JEFFREY et al., 2000;

SISKOVA et al., 2009). Intact synapses have pre-synaptic terminals packed with electron-

lucent cytoplasm, characteristic small round synaptic vesicles and opposing bar-like post-

synaptic densities. Degenerating synapses in prion disease are characterized by the presence

of electron dense pre-synaptic terminals, the loss of integrity of vesicles and other organelles.

The pre-synaptic membrane remains intact and the post-synaptic membrane appears to be

increased in the curvature and thickness. With the progression of the disease the post-synaptic

membrane progressively curves around degenerating pre-synaptic elements (SISKOVA et al.,

2009). In advanced stages of prion disease the pre-synaptic terminal appears to be completely

engulfed by a post synaptic density (SISKOVA et al., 2009).

1.6.1. Mechanisms of synaptic dysfunction

Apart from the obvious significance of synapse degeneration in neurodegenerative diseases

extremely little is known about the basic cellular and molecular events by which a misfolded

protein leads to synapse degeneration or dysfunction. Neuronal cell loss, spongiform

appearance and gliosis are prime features of prion diseases; however, the first noticeable

changes emerge to be related to synaptic dysfunction (JEFFREY et al., 2000; SISKOVA et

al., 2009; REIS et al., 2015). In the murine ME7 model of prion disease, early behavioral

deficits emerge in conjunction with PrPSc

deposition and synaptic dysfunction preceding

neuronal death (JEFFREY et al., 2000; RUSSELAKIS-CARNEIRO et al., 2004; SOTO and

SATANI, 2010). Studies performed on knock-out mice have also highlighted the role of

prion proteins in synaptic function (COLLINGE et al., 1994). PrPC enriched in the synapses

interacts with proteins participating in synaptic transmission e.g. synaptophysin (FOURNIER

22 Chapter 1

et al., 1995; HAEBERLE et al., 2000; BROWN, 2001). Immunohistologically, abnormal

PrPSc

staining is found in the region of neuronal cell bodies and dendrites, mimicking

synaptophysin distribution, also signifying abnormal PrPSc

accumulation in synaptic

structures (KITAMOTO et al., 1992; FOURNIER et al., 1995). During the initial stages of

the disease, PrPSc

accumulates in membrane lipid rafts. This accumulation leads to the

detachment of caveolin and synaptophysin from these membrane domains and probably

impacts synaptic function (RUSSELAKIS-CARNEIRO et al., 2004). Exocytosis and

neurotransmission linked proteins e.g. SNAP-25, syntaxins, synaptophysin cysteine string

protein (CSP), VAMP-2, synapsin and Rab3a have also been reported to be decreased in

prion disease in the CNS (FERRER et al., 2000; GRAY et al., 2009; HILTON et al., 2013).

Biochemical analysis highlighted the fact that loss of synaptic vesicle proteins, especially

CSP, VAMP-2, and synapsin precedes the changes of proteins in the post-synaptic division

(GRAY et al., 2009). CSPα, as an important synaptic protein, exists in pre-synaptic terminals

and forms a chaperone complex to maintain normal synapses (TOBABEN et al., 2001). It is

of particular interest that mice lacking CSP demonstrate a synaptic degenerative phenotype

(FERNÁNDEZ-CHACÓN et al., 2004). Depletion or reduction of CSPα results in an

abnormal SNAP-25 conformation that resists soluble NSF attachment protein receptor

(SNARE) complex formation, and is subject to ubiquitylation and proteasomal degradation

(SHARMA et al., 2011a; YI and EHLERS, 2007). An impairment of the SNARE complex

due to an alteration or reduction in SNAP-25 finally correlates to neurodegeneration

(SHARMA et al., 2011b; HE et al., 2003). Thus deletion or reduction of CSPα may result in a

massive neurodegeneration at the synaptic level that impairs survival in the ME7 model of

prion disease (FERNÁNDEZ-CHACÓN et al., 2004).

Summarized, these data indicate that conversion of normal PrPC to abnormal PrP

Sc affects the

strength and function of synapses, ultimately leading to neurological damage and finally

initiating the clinical onset of disease (HILTON et al., 2013).

1.6.1.1. Role of mitochondria in synaptic degeneration

Damage or dysfunctions of mitochondria are frequently associated with neurodegenerative

diseases and it is well documented that neuronal synaptic function and mitochondria are co-

dependent (CASTELLANI et al., 2002; LI et al., 2004; SISKOVA et al., 2010). However, the

exact mechanism of mitochondrial contribution to neurodegeneration has not been explored

Chapter 1 23

completely. A recent study in the ME7 model reported that synaptic pathology was

accompanied by alterations in mitochondria (SISKOVA et al., 2010). The phenomenon of

early involvement of neuronal mitochondria is further detailed by the finding that N-acetyl

aspartate (synthesized by neuronal mitochondria) level decreased in the thalamus and

hippocampus as well as in brain areas associated with the early onset of behavioral deficits

(SISKOVA et al., 2010). During the initial stage of prion disease, synaptic density remains

unchanged. This requires decreased respiration which leads to reduction or silencing of

mitochondrial function, followed by a withdrawal of the degenerated synaptic terminal from

the remaining axonal portion. Neuronal mitochondria exhibit various morphological changes

in the inner membrane following prion disease progression. Chronologically, succinate

dehydrogenase and cytochrome c oxidase (COX) activity assays showed structural changes in

mitochondria with functional impairment of complex IV activity in the initial stage of prion

disease. Impairment of complex IV activity leads to compromised mitochondrial respiratory

activity in prion disease and corresponds with the beginning of synaptic loss (SISKOVA et

al., 2010). In addition, up-regulation of nitric oxide can be accompanied by astrogliosis in

prion diseases (ALMER et al., 1999; GRAY et al., 2009). It is predicted that nitric oxide is an

effective mediator of brain damage and may directly disturb mitochondrial function by

interfering to the oxygen binding to complex IV (CLEETER et al., 1994; BOLANOS et al.,

1997; SISKOVA et al., 2010). In addition expression of nitric oxide may disturb

mitochondrial respiratory chain complex I and IV activity (SMITH and LASSMANN, 2002;

ZHANG et al., 2005; SISKOVA et al., 2010). During the initial stage of prion disease, a

significant increase in neuronal nitric oxide synthase in the hippocampus has been observed

(PICANCO-DINIZ et al., 2004; SISKOVA et al., 2010). The increased production of

neuronal nitric oxide takes place in the stratum radiatum and a decline in the late disease

stage are paralleling COX activity changes and hence justifying the idea of nitric oxide

involvement in the damage of mitochondria in the ME7 scrapie model (SISKOVA et al.,

2010). In ultrastructural studies of the ME7 murine model, neuronal mitochondria appeared

reduced in number, swollen, having significantly large diameter and poorly defined swollen

cristae as compared to healthy wild type mice (SISKOVA et al., 2010). Due to the presence

of morphological defects and complex IV activity dysfunction, the respiratory capability of

neuronal mitochondria in prion disease could be compromised in the initial stage of the prion

disease and may correspond to the initiation of synaptic dysfunction. A misbalance in

24 Chapter 1

reactive oxygen levels, along with other changes, could be induced and may contribute to the

intensification of neuropathological processes (SISKOVA et al., 2010).

1.7. Aims of the study

It is well-known that the gene encoding the prion protein (PRNP) critically influences the

susceptibility of small ruminants for certain forms of TSEs, which has contributed to the

development of selective breeding programs, for instance of sheep with a lower susceptibility

to scrapie (AGUILAR-CALVO et al., 2014). Moreover, transgenic mice expressing a certain

polymorphic variant of the goat PRNP gene are resistant to scrapie and BSE (AGUILAR-

CALVO et al., 2014). However, whether the genotype similarly has an impact on the

susceptibility of goats for BSE remained enigmatic so far. A detailed elucidation of this

question has important implications for the control of TSEs as it will contribute to identify

appropriate genotypes, which could selectively be chosen for targeted breeding programs of

goats.

While abundant data exist, which contributed to a detailed insight into the neuropathogenesis

of TSEs in the brain itself, comparatively less is known about the involvement of the

vegetative nervous system and the spinal cord, even though the sympathetic nervous system

has been demonstrated to play a pivotal role in the initial spread of prions in BSE-infected

cattle. Research upon BSE has mainly focused on cattle; however, the pathogenesis in goats

has been subjected to little research so far. Moreover, the cellular and molecular constituents

and mechanisms that facilitate the spread of the agent remain undetermined.

Thus, the present study aimed to (i) clarify the effect of certain PRNP genotypes in the oral

transmission of the BSE agent to goats, (ii) to detect axonal cytoskeletal and transport

disturbances during the course of BSE in the spinal cord and peripheral tissues of

experimentally infected goats, and lastly (iii) to unravel potential ultrastructural changes in

the superior cervical ganglion of experimentally BSE-infected goats with a special emphasis

upon synapse pathology.

Chapter 2 25

Chapter 2

26 Chapter 2

Chapter 2 EFFECT of Q211 and K222 PRNP

POLYMORPHIC VARIANTS IN THE

SUSCEPTIBILTY OF GOATS TO ORAL

INFECTION WITH GOAT BOVINE

SPONGIFORM ENCEPHALOPATHY

AGUILAR-CALVO, P., FAST, C., TAUSCHER, K., ESPINOSA, J-C., GROSCHUP, M. H.,

NADEEM, M., GOLDMANN, W., LANGEVELD, J., BOSSERS, A., ANDREOLETTI, O.

TORRES, J-M.

BACKGROUND: The prion protein-encoding gene (PRNP) is one of the major

determinants for scrapie occurrence in sheep and goats. However, its effect on bovine

spongiform encephalopathy (BSE) transmission to goats is not clear.

METHODS: Goats harboring wild-type, R/Q211 or Q/K222 PRNP genotypes were orally

inoculated with a goat-BSE isolate to assess their relative susceptibility to BSE infection.

Goats were killed at different time points during the incubation period and after the onset of

clinical signs, and their brains as well as several peripheral tissues were analyzed for the

accumulation of pathological prion protein (PrPSc

) and prion infectivity by mouse bioassay.

RESULTS: R/Q211 goats displayed delayed clinical signs compared with wild-type goats.

Deposits of PrPSc

were detected only in brain, whereas infectivity was present in peripheral

tissues too. In contrast, none of the Q/K222 goats showed any evidence of clinical prion

disease. No PrPSc

accumulation was observed in their brains or peripheral tissues, but very

low infectivity was detected in some tissues very long after inoculation (44-45 months)

CONCLUSIONS: These results demonstrate that transmission of goat BSE is genotype

dependent, and they highlight the pivotal protective effect of the K222 PRNP variant in the

oral susceptibility of goats to BSE.

KEYWORDS: BSE; PRNP polymorphisms; goats; susceptibility/resistance; transgenic mice

Published in Journal of infectious diseases 2015, DOI: 10.1093/infdis/jiv112.

Chapter 2 27

AUTHORS CONTRIBUTIONS

AGUILAR-CALVO, P., FAST, C., TAUSCHER, K., ESPINOSA, J-C., GROSCHUP, M. H.,

NADEEM, M., GOLDMANN, W., LANGEVELD, J., BOSSERS, A., ANDREOLETTI, O.

TORRES, J-M.

EFFECT of Q211 and K222 PRNP POLYMORPHIC VARIANTS IN THE SUSCEPTIBILTY

OF GOATS TO ORAL INFECTION WITH GOAT BOVINE SPONGIFORM

ENCEPHALOPATHY.

Published in The Journal of Infectious Diseases 2015, DOI: 10.1093/infdis/jiv112.

P Aguilar-Calvo and J.-C. Espinosa were involved in the study design, coordination of the

mouse bioassays, and drafted the manuscript.

C. Fast and K. Tauscher were involved in the study design, coordination of the goat

experiments, and drafted the manuscript.

M. H. Groschup was involved in the study design, the coordination of the goat experiments,

editing the manuscript, and in obtaining funding.

M. Nadeem has performed the immunohistochemistry and analyzed the data.

W. Goldmann was involved in the coordination of the study and in editing the manuscript.

J. Langeveld and A. Bossers were involved in in the study design, funding obtainment, and in

editing the manuscript.

O. Andreoletti was involved in the coordination of the study and in editing the manuscript.

J.-M. Torres was involved in the coordination of the animal experiments, coordination of

mouse bioassays, obtained funding, and edited the manuscript

28 Chapter 2

Chapter 3 29

Chapter 3

30 Chapter 3

Chapter 3 BSE INFECTION OF GOATS ALTERS

NEUROFILAMENT

PHOSPHORYLATION STATUS OF

SPINAL CORD AXONS

NADEEM, M., SPITZBARTH, I., HAIST, V., ROHN, K., TAUSCHER, K., ROHN, K.,

BOSSER, A., LANGEVELD, J., GROSCHUP, M.H., BAUMGÄRTNER, W., FAST, C.,

GERHAUSER, I.

Abstract

Transmissible spongiform encephalopathies (TSEs) represent a group of progressive diseases

that affect the nervous system of humans and various animal species. Recently reported

bovine spongiform encephalopathy (BSE) infections in goats in the United Kingdom and

France have brought small ruminant species into the focus of prion disease research.

Immunohistochemistry was performed to detect axonal cytoskeletal and transport

disturbances during the course of BSE in the spinal cord and autonomous ganglia of

experimentally infected goats. Interestingly, the present study demonstrated abnormal

expression of non-phosophorylated neurofilament (nNF) in axons of the white matter of the

spinal cord, which was restricted to goats with clinical BSE. The results indicate axonal

damage and disturbances in axonal transport during BSE in goats. However, whether

abnormal nNF accumulations are related to disturbed axonal transport mechanisms in TSEs,

the spread of TSE agents, and neuronal degeneration, has to be evaluated in future studies.

Moreover, the study reports for the first time that there is immunohistochemical evidence for

PrPSc

deposition in spinal cord white matter glial cells of BSE positive goats, indicating the

involvement of glial cells in the spread of the agent.

Submitted for publication

Chapter 3 31

AUTHORS CONTRIBUTIONS

NADEEM, M., SPITZBARTH, I., HAIST, V., ROHN, K., TAUSCHER, K., ROHN, K.,

BOSSER, A., LANGEVELD, J., GROSCHUP, M.H., BAUMGÄRTNER, W., FAST, C.,

GERHAUSER, I.

BSE INFECTION OF GOATS ALTERS NEUROFILAMENT PHOSPHORYLATION

STATUS OF SPINAL CORD AXONS.

Submitted for publication.

M. Nadeem performed immunohistochemistry and electron microscopy, analyzed the

data, and drafted the manuscript.

I. Spitzbarth was involved in the coordination of the immunohistochemical studies,

performed statistical analysis, and drafted the manuscript.

V. Haist was involved in the coordination of the ultrastructural studies and revised the

manuscript.

K. Rohn was involved in performing electron microscopy and generation of ultrastructural

photographs.

K. Tauscher performed the animal experiments and obtained tissue samples.

K. Rohn was involved in the statistical analysis of the data.

A. Bosser, J. Langeveld, and M. H. Groschup were involved in the study design, the

coordination of the animal experiments, revision of the manuscript, and in obtaining funding.

W. Baumgärtner was involved in the coordination of the immunohistochemical and

ultrastructural studies, edited the manuscript, and obtained parts of the funding.

C. Fast was involved in the study design, coordination of the animal experiments, the

conduction of immunohistochemistry, and edited the manuscript.

I. Gerhauser was involved in the coordination of the immunohistochemical studies,

performed statistical analyses, designed figures, and edited the manuscript.

32 Chapter 3

SHORT COMMUNICATION

BSE infection of goats alters the neurofilament phosphorylation status of spinal cord

axons

Muhammad Nadeem1,2,*

, Ingo Spitzbarth1,2,*

, Verena Haist1,

Kerstin Rohn1, Kerstin

Tauscher3, Karl Rohn

4, Alex Bossers

5, Jan Langeveld

5, Martin H. Groschup

3, Wolfgang

Baumgärtner1,2,#

, Christine Fast

3,+, Ingo Gerhauser

1,+

1 Department of Pathology, University of Veterinary Medicine, Hannover, Germany

2 Center for Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany

3 Friedrich Loeffler Institute, Institute of Novel and Emerging Infectious Diseases,

Greifswald-Insel Riems, Germany

4 Department of Biometry, Epidemiology and Information Processing, University of

Veterinary Medicine, Hannover, Germany

5 Central Veterinary Institute, Wageningen UR, Lelystad, The Netherlands

* both authors contributed equally to this study and should be considered co-first authors

(first authors in alphabetical order)

+ both authors contributed equally to this study and should be considered co-last authors (last

authors in alphabetical order)

# Corresponding author:

Prof. Dr. Wolfgang Baumgärtner, Ph.D.

University of Veterinary Medicine Hannover, Department of Pathology

Bünteweg 17, D-30559 Hannover, Germany

Tel.: +49-(0)-511-953-8620; Fax: +49-(0)-511-953-8675

E-mail: [email protected]

Chapter 3 33

TRANSMISSIBLE SPONGIFORM ENCEPHALOPATHIES (TSEs) including bovine

spongiform encephalopathy (BSE) are devastating neurodegenerative disorders caused by

conversion of the normal cellular prion protein (PrPC) into an abnormal isoform (PrP

Sc;

Prusiner, 1982; Chesebro, 2003). Following the discovery of two goat BSE cases in the UK

and France (Eloit and others 2005; Jeffery and others 2006) small ruminants were considered

to pose an BSE infection risk/source for cattle and humans in particular. In goats

experimental TSE susceptibility strongly depends on polymorphisms in the prion protein

gene (PRNP) (Aguilar-Calvo and others 2014; Aguilar-Calvo and others 2015). In particular,

goats with R/Q211 polymorphism (IQQ/IRQ) and Q/K222 polymorphism (IRK/IRQ) show

delayed or even absent clinical signs compared with wild-type goats (IRQ/IRQ) (Aguilar-

Calvo and others 2015). After oral infection of ruminants, the agent spreads via the

autonomous nervous system, ultimately resulting in manifest disease in the brain (Van

Keulen and others 2002; Hoffmann and others 2007; Kaatz and others 2012). However, the

cellular and molecular mechanisms that facilitate the spread of prions within the nervous

system and the involvement of the spinal cord in the pathogenesis of TSEs has been subjected

to little research so far, even though PrPSc

has been detected in the spinal cord (Flechsig and

others 2000; Fukuda and others 2012; Kaatz and others 2012).

The aim of the present study was to elucidate potential changes in the antigen expression of

various immunohistochemical markers in regions of the sympathetic infection route, which

have been demonstrated to represent key localizations for the spread of the BSE agent (Kaatz

and others 2012), including the celiac and mesenteric ganglion complex (CMGC) and the

spinal cord. Moreover, we sought to characterize ultrastructural changes in neurons of the

superior cervical ganglion (SCG).

Samples were collected during an oral BSE challenge (1g BSE-positive homogenized caprine

brain) and serial kill study of Alpine-Saanen-mixed breed goats approved by the local

authorities of the Federal State of Mecklenburg-Western Pomerania, Germany (LALLF

7221.3-2.5-001/05). The goats carried different PRNP genotypes with differing susceptibility.

Parts of the same population have been described in a previously published study (Aguilar-

Calvo and others, 2015; table 1). The animals were killed 6-45 months post infection (mpi;

table 1). Formalin-fixed tissue samples were incubated for 1 h in 98% formic acid prior to

paraffin embedding (Kaatz and others 2012). Three-micrometer sections were stained with

haematoxylin and eosin (HE).

34 Chapter 3

Table 1: Summary of the animals used in the present study, their clinical BSE status

and results of immunohistochemistry for abnormal prion protein in superior cervical

ganglia, celiac and mesenteric ganglion complex, and spinal cord of controls, BSE-

infected goats, and their offspring (goat kids)

Genotype

Animal

ID

Animal

consecutive

number

Gender Necropsy

(months

post

infection)

Clinical

status*

PrPSc

(IHC)**

SCG

PrPSc

(IHC)**

CGMC

PrPSc

(IHC)**

Spinal cord

IRQ/IRQ

ZG 26 1 m 6 - n.d. - -

ZG 32 2 m 6 - - - -

ZG 35 3 m 6 - - - -

ZG 19 4 m 12 - - - -

ZG 24 5 m 12 - - - -

ZG 30 6 m 12 - n.d. - -

ZG 08 7 m 12 - - - -

ZG 13 8 m 24 - - - -

ZG 01 9 f 24 +++ n.d. + +

ZG33 10 m 24 + + + +

ZG 34 11 f 25 +++ n.d. + +

ZG38 12 m 25 +++ + n.d. +

ZG 39 13 m 25 - n.d. - -

ZG 18 14 f 46 - n.d. - -

IQQ/IRQ

ZG 04 15 m 6 - - - -

ZG 06 16 f 6 - n.d. - -

ZG 36 17 m 6 - - - -

ZG 21 18 m 9 - - - -

ZG 22 19 m 12 - - - -

ZG 31 20 m 12 - - - -

ZG 28 21 m 33 +++ + n.d. +

ZG 05 22 f 34 ++ n.d. + +

ZG 12 23 f 34 - n.d. - -

ZG 20 24 f 36 ++ n.d. + +

IRK/IRQ

ZG 16 25 m 6 - - - -

ZG 23 26 m 6 - n.d. - -

ZG 37 27 m 6 - - - n.d.

ZG 02 28 m 12 - - - -

ZG 09 29 m 12 - - - n.d.

ZG 10 30 m 24 - - - -

ZG 15 31 f 36 - n.d. - -

ZG 25 32 f 44 - n.d. - -

ZG 11 33 f 45 - - - -

Controls

(IRQ/IRQ)

Kz-ISG

1 34 m 12 - - - -

Kz-ISG

2 35 m 12 - - - -

Horizontal

controls

(different

genotypes)

ZG 27 36 m 14 - - - -

ZG 14 37 m 31 - - - -

ZG 40 38 m 53 - - - -

Goat kids

(different

genotypes)

ZG 42 39 -- 27*** - n.d. - -

ZG 44 40 -- 26*** - n.d. - -

ZG 45 41 -- 26*** - n.d. - -

ZG 46 42 -- 13*** - n.d. n.d. -

ZG 47 43 -- 25*** - n.d. n.d. -

Chapter 3 35

m = male, f = female, IHC = immunohistochemistry, SCG = superior cervical ganglion, CMGC = celiac and

mesenteric ganglion complex, n.d. = not determined

* - = no clinical signs; + = mild subclinical signs, ++, +++ = moderate to severe clinical signs;

** - = negative in immunohistochemistry for PrPSc

; + = positive in immunohistochemistry for PrPSc

;

*** = age of the animals (not infected)

PrPSc

was visualized using a monoclonal primary antibody as described (clone 6C2; Central

Veterinary Institute, Wageningen UR, Lelystad, Netherlands; Kaatz and others 2012).

Immunohistochemistry was additionally performed on CMGC and spinal cord sections using

markers for microtubules, axonal motor proteins, axonal damage, intermediate filaments,

peripheral and central myelin proteins, and antigen presenting cells as described (Seehusen

and Baumgärtner, 2010; Bock and others 2013; Spitzbarth and others 2011; table 2). SCG

samples were processed for ultrastructural analyses as described (Baumgärtner and others

1987; Ulrich and others 2008).

Table 2: Summary of the antibodies used to characterize neurons and satellite glial cells

of the celiac and mesenteric ganglion complex in controls and BSE infected goats using

immunohistochemistry

Detected

antigen Clonality

Primary antibody

(Supplier) Pretreatment

Blocking

solution

Primary

antibody

dilution

PrPSc

Mouse mc

Clone 6C2, Jan Langeveld,

CIDC Lelystad,

Netherlands

Citric

buffer/autoclave/20' - 1.50

alpha-

acetylated

Tubulin

Mouse mc Sigma T 6793 Citric buffer/MW/20' Horse

serum 1:5 1:1500

beta-APP Mouse mc Chemicon M348 Lot

LV1622798 Citric buffer/MW/20'

Horse

serum 1:5 1:1250

CNPase Mouse mc Millipore MAB 326 Lot

LV 1567855 Citric buffer/MW/20'

Horse

serum 1:5 1:500

GFAP Rabbit pc DAKO Z 0334 Lot

23120676 -

Horse

serum 1:5 1:6000

Iba-1 Rabbit pc Wako 019-19741 Citric buffer/MW/20' Horse

serum 1:5 1:500

Kinesin 5 A Rabbit pc Sigma K 0889 Citric buffer/MW/20' Horse

serum 1:5 1:400

36 Chapter 3

MAP-2 Mouse mc Chemicon, MAB 3418 Citric buffer/MW/20' Horse

serum 1:5 1:500

MHC II Mouse mc

DAKO M0746 HLA-DR

Clone TAL.1B5 Lot

00049249

Citric buffer/MW/20' Horse

serum 1:5 1:40

n-NF Mouse mc Sternberger Monoclonals

Inc. SMI 311 Lot 9 Citric buffer/MW/20'

Horse

serum 1:5 1:1000

p75NTR

Mouse mc Hybridoma, Clone HB-

8737, ATCC, USA -

Horse

serum 1:5 1:5

Periaxin Rabbit pc Sigma HPA001868 Citric buffer/MW/20' Horse

serum 1:5 1:2500

p-NF Mouse mc Sternberger Monoclonals

Inc. SMI 312 Lot 9

Triton-X 0,25% in

PBS 15'

Horse

serum 1:5 1:8000

Synaptophysin Mouse mc DAKO SY38 Citric buffer/MW/20' Horse

serum 1:5 1:50

Tau-1 Mouse mc Chemicon MAB 3420 Citric buffer/MW/20' Horse

serum 1:5 1:2000

Vimentin Mouse mc DAKO M 0725 Lot

00051105 -

Horse

serum 1:5 1:75

βIII TubulinIII Mouse mc Sigma T 8660 Citric buffer/MW/20' Horse

serum 1:5 1:8000

20' = 20 minutes; beta APP = beta amyloid precursor protein; CNPase = 2',3'-cyclic-nucleotide 3'-

phosphodiesterase; GAP 43 = growth-associated protein 43; GFAP = glial fibrillar associated protein; Iba-1 =

ionized calcium-binding adapter molecule 1; mc = monoclonal; MAP-2 = microtubules associated protein 2;

MHC II = major histocompatibility complex class II; MW = microwave; n-NF = non-phosphorylated

neurofilament; p75NTR

= low affinity neurotrophin receptor p75; pc = polyclonal; p-NF = phosphorylated

neurofilament

Clinical signs typical for BSE were detected in three IRQ/IRQ goats (animal numbers: 9, 11,

12) and three IQQ/IRQ goats (animal numbers: 21, 22, 24), which developed

hypersensitivity, weight loss, lethargy, and alopecia at advanced time points beginning at 24

mpi (Aguilar-Calvo and others 2015; table 1). In the spinal cord, BSE-typical vacuolation of

the neuropil and PrPSc

-deposition were evident in all clinical animals and one subclinical goat

(case number: 10). Besides the grey matter, PrPSc

-positivity was observed in glial cells but

not in axons of the spinal cord white matter in all 7 positive goats (table 3). Despite the lack

of histological lesions PrPSc

was detected in the SCG of three animals (case numbers: 10, 12,

and 21). Unfortunately, SCG tissue was not available from some animals including the 4

remaining PrPSc

-positive goats (table 1). In the CMGC, 5 animals displayed PrPSc

-

Chapter 3 37

immunopositivity (case numbers: 9, 10, 11, 22, and 24) and three of them also neuronal

vacuolation (case numbers: 9, 11, and 22). No CGMC tissue was available in goats 12 and 21

(table 1). Strikingly, expression of non-phosphorylated neurofilament (n-NF) was not

detected in any spinal cord axon of the PrPSc

-negative animals, but was significantly up-

regulated in both swollen and normal appearing axons of the spinal cord white matter in 6

PrPSc

-positive animals (figure 1). In contrast, expression of all other investigated antigens

(table 2) was not influenced by the BSE status. Similarly, synapses were normal in SCG

neurons in both control and PrPSc

-positive goats at the ultrastructural level. Mitochondrial

swelling, cristolysis, and occasionally dilatation of rough endoplasmic reticulum was noted in

animals of each genotype, PrPSc

-status, and age, thus most probably indicating post-mortem

artifacts.

Table 3: Summary of the deposition of prion protein in spinal cord grey and white

matter of BSE-positive goats

Groups

(variants

of prion

genes)

Animal

consecutive

number

Animal

internal

number

PrPSc

deposition

grey matter

(IHC)

PrPSc

deposition white matter

(IHC)

Neuronal

vacuolation

ST AT DT Axons

IRQ/IRQ 9 ZG 01 +++ +++ +++ +++ - +++*

IRQ/IRQ 10 ZG 33 ++ + - - - +

IRQ/IRQ 11 ZG 34 +++ ++ ++ + - +

IRQ/IRQ 12 ZG 38 ++ + + + - +

IQQ/IRQ 21 ZG 28 +++ +++ +++ +++ - +

IQQ/IRQ 22 ZG 05 +++ + + + - ++*

IQQ/IRQ 24 ZG 20 +++ + - - - +++*

ST = spinal tract, AT = ascending tract, DT = descending tract,* = spongiform changes

+ - +++ mild to severe immunopositivity

The present study demonstrates for the first time that BSE infection of goats is associated

with glial PrPSc

-deposition in the spinal cord white matter along with unsuspected alterations

in the neurofilament phosphorylation status of axons. While absent in normal axons,

abnormal neurofilament expression has been shown in various neurologic diseases including

Creutzfeldt-Jakob disease (Gajdusek, 1985; Liberski and Budka, 1999; Tsunoda and

Fujinami, 2002; Seehusen and Baumgärtner, 2010; Bock and others 2013). Axonal

expression of n-NF in the present study is indicative of disturbances in axonal transport

processes. Underlining this hypothesis, some immunopositive axons had an increased

diameter (figure 1).

38 Chapter 3

Figure 1: Immunohistochemical demonstration of non-phosphorylated neurofilament

(n-NF) in axons of a control animal and a goat with experimental infection with bovine

spongiform encephalopathy (BSE).

A: Spinal cord from a control animal with absent expression of n-NF in axons of the white

matter of the spinal cord. B: Spinal cord of a BSE infected goat revealing increased axonal

expression of n-NF in both axons with a normal appearing diameter (arrows) and swollen

axons (spheroids, arrowhead) at > 24 months post infection, indicating axonal damage with

disrupted axonal transport processes. Immunohistochemistry with the avidin-biotin-

peroxidase-complex method. Chromogen: 3,3'-diaminobenzidine. Bars = 20 μm.

Synaptic disorganization and loss has been indicated as a constant feature of prion disease

(Clinton and others 1993; Jeffrey and others 2000). Nevertheless, ultrastructural lesions were

lacking in the SCG of PrPSc

-positive goats in the present study. Consequently, BSE-induced

synaptic pathology in goats might be restricted to the brain itself, which has to be confirmed

in future investigations. The lack of PrPSc

-deposition in IRK/IRQ animals substantiates their

low susceptibility to clinical BSE (Aguilar-Calvo and others 2015). In contrast to earlier

reports in cattle (Kaatz and others, 2012), PrPSc

-deposition was not detected in peripheral

ganglia prior to involvement of the spinal cord (table 1). However, ganglia were not available

from all animals excluding definitive conclusions on the time course of the infection. In

summary, our findings suggest that axonal damage and disturbed axonal transport in the

spinal cord white matter might play a so far underestimated role in the pathogenesis of TSEs.

Chapter 3 39

Acknowledgements

This study was in part supported by the Niedersachsen-Research Network on

Neuroinfectiology (N-RENNT) of the Ministry of Science and Culture of Lower Saxony and

in part by European Union projects (FOOD-CT-2006-36353, GoatBSE, and 219235 ERA-

NET EMIDA, GOAT-TSE-FREE). Muhammad Nadeem is thankful to Higher Education

Commission (HEC), Pakistan and Deutscher Akademischer Austauschdienst (DAAD),

Germany for providing a scholarship. The authors acknowledge the work of Dr. Susanne

Niedermeyer, Gesine Kreplin, Bettina Buck, Petra Grünig, Claudia Herrmann, Christiane

Namneck, Kerstin Schöne, Caroline Schütz, and Brigitte Behrens.

References

AGUILAR-CALVO, P., ESPINOSA, J.C., PINTADO, B., GUTIÉRREZ-ADÁN, A.,

ALAMILLO, E., MIRANDA, A., PRIETO, I., BOSSERS, A., ANDREOLETTI, O.

& TORRES, J.M. (2014) Role of the goat K222-PrPC polymorphic variant in prion

infection resistance. Journal of Virology 88, 2670-2676

AGUILAR-CALVO, P., FAST, C., TAUSCHER, K., ESPINOSA, J-C., GROSCHUP, M. H.,

NADEEM, M., GOLDMANN, W., LANGEVELD, J., BOSSERS, A.,

ANDREOLETTI, O. & TORRES, J-M. (2015) Effect of Q211 and K222PRNP

polymorphic variants in the susceptibility of goats to oral infection with Goat-BSE.

The Journal of Infectious Diseases 10, jiv112

BAUMGÄRTNER, W., KRAKOWKA, S. & BLAKESLEE, Z.R. (1987) Persistent infection

of Vero cells by paramyxoviruses a morphological and immunoelectron microscopic

investigation. Intervirology 27, 218-223

BOCK, P., SPITZBARTH, I., HAIST, V., STEIN, V.M., TIPOLD, A., PUFF, C., BEINEKE,

A. & BAUMGÄRTNER, W. (2013) Spatio-temporal development of axonopathy in

canine intervertebral disc disease as a translational large animal model for

nonexperimental spinal cord injury. Brain Pathology 23, 82-99

CHESEBRO, B. (2003) Introduction to the transmissible spongiform encephalopathies or

prion disease. British Medical Bulletin 66, 1-20

CLINTON, J., FORSYTH, C., ROYSTON, M.C. & ROBERTS, G.W. (1993) Synaptic

degeneration is the primary neuropathological feature in prion disease: a preliminary

study. Neuroreport 4, 65-68

40 Chapter 3

ELOIT, M., ADJOU, K., COULPIER, M., FONTAINE, J.J., HAMEL, R., LILIN, T.,

MESSIAEN, S., ANDREOLETTI, O., BARON, T., BENCSIK, A., BIACABE, A.G.,

BERINGUE, V., LAUDE, H., LE DUR, A., Vilotte, J.L., COMOY, E., DESLYS,

J.P., GRASSI, J., SIMON, S., LANTIER, F. & SARRADIN, P. (2005) BSE agent

signatures in a goat. The Veterinary Record 16, 523-524

FLECHSIG, E.D., SHMERLING, I., HEGYI, A., RAEBER, J., FISCHER, M., FISCHER,

M., COZZIO, A., von MERING, C., AGUZZI, A. & WEISSMANN C (2000) Prion

protein devoid of the octapeptide repeat region restores susceptibility to scrapie in PrP

knockout mice. Neuron 27, 399-408

FUKUDA, S., ONOE, S., NIKAIDO, S., FUJII, K., KAGEYAMA, S., IWAMARU, Y.,

IMAMURA, M., MASUJIN, K., MATSUURA, Y., SHIMIZU, Y., KASAI, K.,

YOSHIOKA, M., MURAYAMA, Y., MOHRI, S., YOKOYAMA, T. & OKADA, H.

(2012) Neuroanatomical distribution of disease-associated prion protein in

experimental bovine spongiform encephalopathy in cattle after intracerebral

inoculation. The Journal of Infectious Diseases 65, 37-44

GAJDUSEK, D.C. (1985) Hypothesis: interference with axonal transport of neurofilaments

as a common pathogenetic mechanism in certain diseases of the central nervous

system. New England Journal of Medicine 312, 714-719

HOFFMANN, C., ZIEGLER, U., BUSCHMANN, A., WEBER, A., KUPFER, L.,

OELSCHLEGEL, A., HAMMERSCHMIDT, B. & GROSCHUP, M.H. (2007) Prions

spread via the autonomic nervous system from the gut to the central nervous system in

cattle incubating bovine spongiform encephalopathy. Journal of General Virology 88,

1048-1055

JEFFREY, M., HALLIDAY, W.G., BELL, J., JOHNSTON, A.R., MACLEOD, N.K.,

INGHAM, C., SAYERS, A.R., BROWN, D.A. & FRASER, J.R. (2000) Synapse loss

associated with abnormal PrP precedes neuronal degeneration in the scrapie-infected

murine hippocampus. Neuropathology and Applied Neurobiology 26, 41-54

KAATZ, M., FAST, C., ZIEGLER, U., BALKEMA-BUSCHMANN, A.,

HAMMERSCHMIDT, B., KELLER, M., OELSCHLEGEL, A., MCINTYRE, L. &

GROSCHUP, M.H. (2012) Spread of classical BSE prions from the gut via the

peripheral nervous system to the Brain. Immunopathology and Infectious Diseases

181, 515-524

Chapter 3 41

LIBERSKI, P.P. & BUDKA, H. (1999) Neuroaxonal pathology in Creutzfeldt-Jakob disease.

Acta Neuropathologica 97, 329-334

MEDRANO, A.Z., BARMADA, S.J., BIASINI, E. & HARRIS, D.A. (2008) GFP-tagged

mutant prion protein forms intra-axonal aggregates in transgenic mice. Neurobiology

of Disease 31, 20-32

PRUSINER, S.B. (1982) Novel proteinaceous infectious particles cause scrapie. Science 216,

136-144

SEEHUSEN, F. & BAUMGÄRTNER, W. (2010) Axonal pathology and loss precede

demyelination and accompany chronic lesions in a spontaneously occurring animal

model of multiple sclerosis. Brain Pathology 20, 551-559

SPITZBARTH, I., BOCK, P., HAIST, V., STEIN, V.M., TIPOLD, A., WEWETZER, K.,

BAUMGÄRTNER, W. & BEINEKE, A. (2011) Prominent microglial activation in

the early proinflammatory immune response in naturally occurring canine spinal cord

injury. Journal of Neuropathology and Experimental Neurology 70,703-714

TSUNODA, I. & FUJINAMI, R.S. (2002) Inside-Out versus Outside-In models for virus

induced demyelination: axonal damage triggering demyelination. Springer Seminars

in Immunopathology 24, 105-125

ULRICH, R., SEELIGER, F., KREUTZER, M., GERMANN, P.G. & BAUMGÄRTNER, W.

(2008) Limited remyelination in Theiler's murine encephalomyelitis due to

insufficient oligodendroglial differentiation of nerve/glial antigen 2 (NG2)-positive

putative oligodendroglial progenitor cells. Neuropathology and Applied Neurobiology

34, 603-620

van KEULEN, L.J., VROMANS, M.E. & van ZIJDERVELD, F.G. (2002) Early and late

pathogenesis of natural scrapie infection in sheep. APMIS.: Acta pathologica,

microbiologica, et immunologica Scandinavica 110, 23-32

42 Chapter 3

Chapter 4 - Discussion 43

Chapter 4

Discussion

44 Chapter 4 - Discussion

Chapter 4: Discussion

Transmissible spongiform encephalopathies (TSEs) including bovine spongiform

encephalopathy (BSE) and Creutzfeldt-Jakob disease are fatal neurodegenerative diseases

caused by the abnormal isoform (PrPSc

) of the host encoded normal cellular glycoprotein

(PrPC) resulting in motor dysfunctions, dementia, spongiosis, astrogliosis, and neuronal loss

(PRUSINER, 1982; CHESEBRO, 2003; ERMOLAYEV et al., 2009). Though intracerebral

inoculation is the most efficient route for the propagation of TSE agents, the natural route of

exposure appears to be oral (WILLIAMS and MILLER, 2003). Recent studies strongly

support the importance of the lymphoreticular system as well as the peripheral nervous

system (PNS) for neuroinvasion (KLEIN et al., 1997; KÜNZI et al., 2002, KAATZ et al.,

2012). In addition, the sympathetic nervous system seems to play a major role in the initial

neural distribution (KAATZ et al., 2012). In cattle, the sympathetic route of BSE spread

consists of the celiac and mesenteric ganglion complex (CMGC), superior cervical ganglion,

splanchnic nerves, and the intermediolateral cell column of the thoracic spinal cord (KAATZ

et al., 2012). In recent years comprehensive research activities have achieved significant

progress in dissecting the spreading pathways of prion proteins of different TSEs, especially

BSE and scrapie, in naturally as well as in experimentally challenged animals via the oral

route. However, the exact subcellular mechanisms of transport and spread of the agent are

enigmatic so far. A detailed knowledge of prion dissemination in the nervous system might

not only specify new approaches for interfering with prion spread in the PNS, but might also

help to identify therapeutic targets to prevent disease progression.

The onset and distribution of cattle BSE disease specific prion protein accumulations in sheep

and goats are strongly influenced by the genetic predisposition mediated trough the gene

encoding the prion protein (AGUILAR-CALVO et al., 2014; McGOVERN et al., 2015). In

Chapter 4 - Discussion 45

particular, the risk of disease development by prion infection is dependent on the

polymorphism of the prion protein gene (PRNP). This has been observed in sheep with

different PRNP genotypes orally infected with cattle-derived BSE agent (McGOVERN et al.,

2015) and in transgenic mice expressing either the Q222 (wild type) or the K222 variant of goat

PrPC intracerebrally infected with the goat scrapie agent (AGUILAR-CALVO et al., 2014).

AGUILAR-CALVO et al. (2014) described that the K222 PRNP variant might affect PrPC-

PrPSc

interaction and the conversion rate of PrPSc

by the scrapie agent, ultimately resulting in

a delayed incubation period and/or lower susceptibility of Q/K222 compared to wild type-PrP

transgenic mice. The present study aimed to clarify whether the PRNP genotype also affects

the susceptibility of goats to the BSE agent. To achieve this goal, goats harboring different

PRNP genotypes including IRQ/IRQ (Wild type, abbreviated as WT), IQQ/IRQ (abbreviated

as R/Q211), and IRK/IRQ (abbreviated as Q/K222) were orally infected with caprine BSE.

WT goats developed the first clinical signs of TSE 24 months post infection (mpi), which

were consistent with clinical signs previously reported in sheep and goats infected with BSE

(FOSTER et al., 2001a; van KEULEN et al., 2008a). PrPSc

deposits and neurodegenerative

lesions were mainly detected at the level of the obex region in the brain of goats similar to

previous observations in BSE-infected sheep and goats (FOSTER et al., 2001b; GONZÁLEZ

et al., 2005; LEZMI et al., 2011). Prion infectivity, but with lower titers than in the brain, was

also detected in muscles and lymph node tissues, which is consistent with previous findings

in sheep and cattle orally infected with BSE (BUSCHMANN and GROSCHUP, 2005; van

KEULEN et al., 2008b).

In R/Q211 animals, PrPSc

infectivity was higher in brain as compared to peripheral tissue, thus

strongly supporting the results observed in WT goats. However, in contrast to WT goats

R/Q211 animals presented a delayed clinical onset of the disease, starting at 33 mpi, thus

indicating that the PRNP polymorphic variant Q211 might have an influence on the incubation

46 Chapter 4 - Discussion

period of BSE, but not on the general susceptibility. Therefore, R/Q211 polymorphism could

be linked with low resistance against goat BSE compared to the high resistance of this

genotype to scrapie as demonstrated in other field and experimental studies using goats

(BOUZALAS et al., 2010; CORBIÈRE et al., 2013; LACROUX et al., 2014). Consequently,

the prion strain represents a determining factor for TSE occurrence (AGUZZI et al., 2007).

In contrast to the similar susceptibility of WT and R/Q211 goats to goat BSE, none of the

Q/K222 goats examined in this study presented any evidence of disease. In all Q/K222 goats

neither clinical signs nor PrPSc

deposits were observed. Histopathological exanimation also

failed to demonstrate any neuropathological lesions. However, very low infectivity was

detected in the brain of 1 of the 2 Q/K222 goats euthanized at 44-45 mpi, suggesting that K222

PRNP variant drastically decreases the susceptibility of goats to goat BSE.

In cattle, the BSE agent is mainly deposited in the CNS and vegetative nervous system

(BUSCHMANN and GROSCHUP, 2005; KAATZ et al., 2012), whereas it is lymphotropic in

sheep and goats with PrPSc

deposits in several lymphoid tissues (FOSTER et al., 1996;

ANDRÉOLOTTI et al., 2000; BELLWORTHY et al., 2005; KUJALA et al., 2011). In

accordance with this, the present study indicates that the resistance of Q/K222 goats to oral

BSE infection might depend on a lesser capability of goat BSE to alter K222-PrPC in

peripheral tissues. In addition, lesser expression of K222-PrPC

along with a local inconsistency

of the K222-PrPC

isoforms in the structures involved in the major spreading route cannot be

excluded (BERINGUE et al., 2003). In fact, discrepancies in the susceptibility to prion agents

dependent on the inoculation route have been described earlier in R/H154, R/Q211, or Q/K222

goats, which showed complete resistance to the orally administered scrapie agent but were

susceptible to the scrapie agent after intracerebral inoculation (LACROUX et al., 2014).

R/H154 and R/Q211 goats presented 100% susceptibility to intracerebral inoculation of scrapie,

whereas only 2 out of 5 Q/K222 (40%) goats developed clinical disease with 4-5 times longer

Chapter 4 - Discussion 47

incubation periods compared to WT goats, indicating that the K222 variant of PRNP is

associated with less susceptibility of goats to scrapie infection compared to the Q211 variant

(LACROUX et al., 2014). Moreover, our data together with other data from several

experimental (ACUTIS et al., 2012; AGUILAR-CALVO et al., 2014; LACROUX et al.,

2014) and epidemiological studies (ACUTIS et al., 2006; VACCARI et al., 2006;

BARILLET et al., 2009; BOUZALAS et al., 2010; FRAGKIADAKI et al., 2011;

PAPASAVVA-STYLIANOU et al., 2011; ACIN et al., 2013; CORBIÈRE et al., 2013)

emphasize the low susceptibility of the Q/K222 polymorphism to TSE infection.

Consequently, the K222 variant might represent an interesting target to improve breeding

programs in order to control and ultimately eradicate TSE in goats.

The present study also aimed to characterize the role of the spinal cord and autonomic

ganglia in the spread of prion agents in goats. Immunohistochemistry was performed on the

spinal cord and celiac and mesenteric ganglion complex of goats from different genotypic

groups (WT, R/Q211and Q/K222), orally infected with caprine BSE. In addition, synapses were

visualized in the superior cervical ganglion using transmission electron microscopy. WT and

R/Q211 goats exhibited prion protein deposition in the spinal cord, whereas all Q/K222 goats

were negative indicating resistance or lower susceptibility to BSE, and thus substantiating the

previous study (AGUILAR-CALVO et al., 2014). PrPSc

positive goats demonstrated a strong

intra- and perineuronal staining of spinal cord grey matter neurons and glial cells with linear

staining of neuronal processes consistent with previously described prion protein expression

patterns in BSE infection (FUKUDA et al., 2012; VIDAL et al., 2006; JEFFREY and

GONZÁLEZ, 2007). PrPSc

deposition was also observed in spinal cord white matter glial

cells, which was irrespective of ascending or descending tracts, but not in white matter axons.

The present study is the first report describing PrPSc

deposition in spinal cord white matter of

BSE infected goats.

48 Chapter 4 - Discussion

TSEs target neurons and ultimately lead to neuronal loss. However, the role of glial cells in

the uptake, spread, and propagation of prion agents in the CNS during the course of prion

disease has received little attention so far. Previous studies have already suggested that glial

cells, especially astrocytes, may play a key role in the disease process (RAEBER et al., 1997:

MALLUCCI et al., 2003; CRONIER et al., 2004; PRITZKOW et al., 2011, HOLLISTER et

al., 2015). In 263K infected hamsters with clinical signs, abnormal prion agent was detected

in a high proportion of hippocampal astrocytes (YE et al., 1998). In sheep naturally infected

with the scrapie agent, PrPSc

even accumulates to higher levels in astrocytes and other glial

cells than in neurons (van KEULEN et al., 1995; GONZÁLEZ et al., 2002; GONZÁLEZ et

al., 2003). PrPSc

in glial cells has also been observed as early as 8 weeks after intracerebral or

cerebellar inoculation of the scrapie agent in mice, thus suggesting that glial cells, and here

mainly astrocytes, play a pivotal role in the spread of the infection (DIEDRICH et al., 1991).

However, the question how PrPSc

spreads between glial cells and neurons remains unsolved.

Recent in vitro studies of scrapie infected hamster neurons demonstrated that PrPSc

is

transported to late endosomal/lysosomal compartments and throughout cell bodies and

processes of astrocytes and neurons including their contacts (HOLLISTER et al., 2015).

Consequently, glial cells might facilitate the dissemination of PrPSc

towards the CNS and

from the CNS to peripheral tissues.

The PNS seems to be deeply involved in the spread of prion agents from the gut to the CNS

(KAATZ et al., 2012). Consequently, the potential neuropathological effects of goat BSE on

different neuronal, axonal, and satellite glial cell markers was investigated in the celiac and

mesenteric ganglion complex and the spinal cord. However, the PrPSc

status (BSE-positive

vs. BSE-negative goats) did not influence the expression of acetylated α tubulin, βIII tubulin,

β-APP, CNPase, GFAP, Iba1, Kinesin 5A, MAP2, MHC class II, p75NTR

, periaxin,

synaptophysin, pNF, Tau1, and Vimentin by neurons, glial cells, and axons. In addition, the

Chapter 4 - Discussion 49

ultrastructural examination of the superior cervical ganglion did not reveal any significant

changes in cytoplasmic organelles of neurons including mitochondria and the Golgi complex

as well as synapses, which is in contrast to previous findings in CNS lesions of TSEs in mice

(JEFFREY et al., 2000; CUNNINGHAM et al., 2003; SISKOVA et al., 2009). Consequently,

synaptic pathology in goats caused by the BSE agent might be restricted to the CNS itself;

however, this needs to be confirmed in future investigations.

Neurofilaments are a major component of the neuronal cytoskeleton, provide shape and

architecture to axons, and are involved in the relay of signals from the plasma membrane to

the nucleus (TOIVOLA et al., 2005). They are synthesized within neuronal perikarya and

transported to axons via slow axonal transport (MILLER et al., 2002). Within axons

neurofilaments are extensively phosphorylated. Interestingly, the phosphorylation of

neurofilaments seems to be a mechanism for regulating their transport properties, possibly by

promoting their detachment from motor proteins (MILLER et al., 2002). An interference of

neurofilaments with axonal transport has been hypothesized for a long time as a common

pathogenetic mechanism in several CNS diseases (GAJDUSEK, 1985). Similarly,

neuroaxonal pathology, characterized by neuritic swellings and dystrophic neurites seems to

play an important role in TSEs (LIBERSKI and BUDKA, 1999). The present study

documented for the first time that there is abnormal expression of nNF in spinal cord white

matter axons of PrPSc

-positive goats. The cause for the impaired phosphorylation of

neurofilaments yet remains to be elucidated. Nevertheless, nNF accumulations are likely to

compromise axonal transport. However, the influence of nNF on axonal transport

mechanisms and its potential role in the spread of TSE agents as well as neuronal

degeneration needs to be investigated in detail in future studies.

50

Chapter 5 - Summary 51

Chapter 5

Summary

52 Chapter 5 - Summary

Chapter 5: Summary

Susceptibility of goats to the BSE agent with special emphasis on the neuropathogenesis

Muhammad Nadeem

Transmissible spongiform encephalopathies (TSEs) represent a group of progressive diseases

that affect the nervous system of humans and various animal species. The characteristic

neuropathological features of TSEs comprise spongiform changes, astrogliosis, neuronal loss,

and accumulation of an abnormal isoform (PrPSc

) of the host-encoded cellular prion protein

(PrPC). Among animal diseases, bovine spongiform encephalopathy (BSE) is of particular

interest because of its transmissibility to humans causing a variant of Creutzfeldt-Jakob-

disease (vCJD). Recently reported BSE infections in goats in the United Kingdom and France

have brought small ruminant species into the focus of prion disease research. Among TSEs

scrapie affects small ruminants all around the world and its incidence is intensely modified by

the PRNP genotype. In goats several PRNP polymorphisms have been linked to lower

susceptibility to classical scrapie. Among them, the K222 PRNP variant produced most

favorable results, being related with resistance to scrapie in both epidemiological studies as

well in experimental infections in goats and transgenic mice. In contrast, little is known about

the factors defining the risk of goats to BSE.

Studies on orally infected TSEs strongly support the importance of the lymphoreticular

system as well as the peripheral nervous system (PNS) for neuroinvasion. Recent studies in

hamsters, sheep, and cattle demonstrated that TSE agents spread via the sympathetic and/or

parasympathetic nervous system to the central nervous system (CNS). However, the

sympathetic route consisting of the celiac and mesenteric ganglion complex (CMGC), the

superior cervical ganglion, splanchnic nerves, and the intermediolateral cell column of the

Chapter 5 - Summary 53

thoracic spinal cord seems to play the major role in the initial neuronal distribution in orally

BSE-infected cattle. The cellular and molecular constituents and mechanisms that facilitate

the centripetal spread of prions remain undetermined. Principally, prion proteins could use

axonal transport systems, spread cell-free or cell-associated in neural interspaces, sequentially

infect Schwann cells, and convert normal cellular prion protein to abnormal isoforms. The

sequence of events important for the clinical onset of the prion disease and knowledge of the

role of different areas of the CNS and PNS is still fragmentary.

Thus, the present study aimed to clarify the effect of certain PRNP genotypes in the oral

transmission of the goat BSE agent to goats. Immunohistochemistry was performed to detect

axonal cytoskeletal and transport disturbances during the course of BSE in the spinal cord

and peripheral tissues of experimentally infected goats and to elucidate possible pathogenetic

links between ultrastructural changes and PrPSc

accumulation in the superior cervical

ganglion of experimentally BSE-infected goats. Tissues from CNS and PNS from different

genotypes (IRQ/IRQ, IQQ/IRQ and IRK/IRQ) of control and BSE-infected goats were

investigated. IRQ/IRQ wild-type goats presented clinical signs after 24 months post infection

(mpi), while IQQ/IRQ (R/Q211) developed first clinical onset of disease at 33 mpi. In contrast

to the BSE susceptibility of IRQ/IRQ and IQQ/IRQ goats, none of IRK/IRQ (R/K222) goats

presented clinical signs or PrPSc

deposition with neuropathological lesions. However, very

low infectivity was noticed in the brain in 1 of 2 IRK/IRQ goats slaughtered at 44-45 mpi.

The data indicate that IRK/IRQ goats are less susceptible to the transmission of goat BSE

than IRQ/IRQ and IQQ/IRQ, highlighting that the K222 PRNP variant can protect goats

against BSE oral infection. In conclusion, goat BSE agent transmission is genotype-

dependent and the K222 PRNP variant represents a good candidate for better breeding

programs in order to control and eradicate TSEs in goats.

54 Chapter 5 - Summary

The present study did not detect any changes in the expression of various antigens (acetylated

α tubulin, βIII tubulin, β-APP, CNPase, GFAP, Iba1, Kinesin 5A, MAP2, MHC class II,

p75NTR

, periaxin, synaptophysin, pNF, Tau1, and vimentin ) in the spinal cord or the CMGC

of BSE positive compared to negative goats. Moreover, the superior cervical ganglion did not

exhibit ultrastructural changes, which might indicate that synaptic pathology is restricted to

the CNS itself. However, this needs to be confirmed in future studies.

Interestingly, the present study demonstrated abnormal expression of non-phosophorylated

neurofilament (nNF) in axons of the white matter of the spinal cord, which was restricted to

goats with clinical BSE. The results indicate axonal damage and disturbances in axonal

transport during BSE in goats. However, whether abnormal nNF accumulations are related to

disturbed axonal transport mechanisms in TSEs, the spread of TSE agents, and neuronal

degeneration, has to be evaluated in future studies. Moreover, the study reports for the first

time that there is immunohistochemical evidence for PrPSc

deposition in spinal cord white

matter glial cells of BSE positive goats, indicating the involvement of glial cells in the spread

of the agent.

Chapter 6 – Zusammenfassung 55

Chapter 6

Zusammenfassung

56 Chapter 6 - Zusammenfassung

Chapter 6: Zusammenfassung

Die Empfänglichkeit von Ziegen gegenüber BSE unter besonderer Berücksichtigung

der Neuropathogenese

Muhammad Nadeem

Transmissible spongiforme Enzephalopathien (TSEs) stellen eine Gruppe progressiver

Erkrankungen des Nervensystems von Tieren und Menschen dar, deren Neuropathologie

typischerweise durch spongiforme Veränderungen in der grauen Substanz, eine Astrogliose

sowie einen neuronalen Verlust charakterisiert ist. Im Hinblick auf die Pathogenese werden

diese Alterationen durch die Akkumulation einer abnormalen und infektiösen Isoform (PrPSc

)

des von Wirtszellen gebildeten Prionproteins (PrPC) initiiert. Seitdem die Übertragbarkeit der

bovinen spongiformen Enzephalopathie (BSE) auf den Menschen in Form einer Variante der

Creutzfeldt-Jakob-Krankheit (vCJK) bekannt ist, besteht ein großes, öffentliches Interesse an

der Erforschung dieser Erkrankung. Neben dem Rind besteht in der jüngsten Zeit zunehmend

ein wissenschaftliches Interesse an der Erforschung von BSE bei kleinen Wiederkäuern.

Beispielsweise wurde im Vereinigten Königreich und Frankreich eine Infektion von Ziegen

mit BSE nachgewiesen.

Unter den verschiedenen TSEs stellt die Scrapie (Traberkrankheit) die wichtigste Erkrankung

der kleinen Wiederkäuer dar, deren Ausprägung stark vom Genotyp des für das zelluläre

Prionprotein kodierenden Gens (PRNP) abhängt. Bei Ziegen wurden verschiedene PRNP-

Polymorphismen beschrieben, die mit einer differierenden Empfänglichkeit für die klassische

Scrapie einhergehen. Hierbei wiesen mehrere epidemiologische Studien, experimentelle

Infektionen von Ziegen sowie Untersuchungen an transgenen Mäusen auf den positiven

Chapter 6 – Zusammenfassung 57

Einfluss der K222 PRNP-Variante in Bezug auf die Widerstandsfähigkeit gegenüber der

Scrapie-Infektion hin. Dagegen ist der potenzielle Einfluss des PRNP-Genotyps auf eine

BSE-Erkrankung bei Ziegen bisher unbekannt.

Mehrere Studien an oral mit Prionen infizierten Tieren zeigten die große Bedeutung des

lymphoretikulären Systems und des peripheren Nervensystems (PNS) für die Neuroinvasion

des Agens. So gelangen TSE-Erreger bei Hamstern, Schafen und Rindern entlang des

sympathischen und parasympathischen Nervensystems in das zentrale Nervensystem (ZNS).

Bei oral mit BSE infizierten Rindern scheint dem sympathischen Nervensystem die größte

Rolle für die initiale Ausbreitung des Prion-Agens zuzukommen. Hierbei betrifft bei Rindern

eine Prionenausbreitung entlang des sympathischen Nervensystems die Ganglia coeliacum

und mesentericum craniale (GCMC), das Ganglion cervicale craniale, die Nervi splanchnici

und die Substantia intermedia im thorakolumbalen Bereich des Rückenmarks. Die zellulären

und molekularen Komponenten und Mechanismen, die an der zentripetalen Ausbreitung von

Prionen beteiligt sind, sind bisher weitgehend unbekannt. Hypothetisch könnten

Prionenproteine axonale Transportsysteme benutzen, sich im interneuralen Raum zellfrei und

zellgebunden ausbreiten, sequentiell Schwannzellen infizieren, um somit final die

Konformationsänderung des unveränderten zellulären Prionproteins in die abnormale Isoform

zu induzieren. Dennoch bleiben der genaue Zeitverlauf und die Rolle der verschiedenen

Regionen im ZNS und PNS für das klinische Auftreten einer Prionenerkrankung immer noch

unklar.

Die Ziele dieser Studie bestanden darin, den Einfluss bestimmter PRNP-Genotypen auf die

orale Empfänglichkeit für BSE bei Ziegen aufzuklären sowie pathologische Veränderungen

in ausgewählten Lokalisationen des PNS und im Rückenmark zu charakterisieren. Hierfür

wurde eine immunhistologische Untersuchung bei experimentell infizierten Ziegen

durchgeführt, um potenzielle Schäden am axonalen Zytoskelett und Störungen des axonalen

58 Chapter 6 - Zusammenfassung

Transports aufzudecken. Außerdem sollte geklärt werden, inwieweit es einen

pathogenetischen Zusammenhang zwischen ultrastrukturellen Veränderungen und PrPSc

-

Ablagerungen im Ganglion cervicale craniale bei experimentell mit BSE-infizierten Ziegen

gibt. Für diese Untersuchungen wurden verschiedene Gewebe des ZNS und PNS von

Kontrollziegen und BSE-infizierten Ziegen verschiedener Genotypen (IRQ/IRQ, IQQ/IRQ

und IRK/IRQ) entnommen. Die IRQ/IRQ-Wildtyp-Ziegen zeigten die ersten klinischen

Symptome 24 Monate post infectionem (p.i.), während diese bei IQQ/IRQ (R/Q211)-Ziegen

erst 33 Monate p.i. auftraten. Im Gegensatz zu den IRQ/IRQ- und IQQ/IRQ-Ziegen wurden

bei IRK/IRQ (R/K222)-Ziegen weder klinische Symptome noch PrPSc

-Ablagerungen mit

neuropathologischen Veränderungen festgestellt. Allerdings wies das Gehirn einer IRK/IRQ-

Ziege, welche 44 Monate p.i. getötet wurde, eine sehr geringe Infektiosität auf. Diese

Ergebnisse deuten auf eine geringere BSE-Empfänglichkeit von IRK/IRQ-Ziegen gegenüber

IRQ/IRQ- und IQQ/IRQ-Ziegen hin, so dass die K222 PRNP-Variante Ziegen möglicherweise

vor einer oralen BSE-Infektion schützen kann. Somit stellt die gezielte Zucht der K222 PRNP-

Variante möglicherweise eine geeignete Möglichkeit dar, natürliche Fälle von TSE bei

Ziegen auszumerzen.

In der vorliegenden Studie wurde die Expression zahlreicher Antigene (azetyliertes α-

Tubulin, βIII Tubulin, β-APP, CNPase, GFAP, Iba1, Kinesin 5A, MAP2, MHC class II,

p75NTR

, Periaxin, Synaptophysin, pNF, Tau1 und Vimentin) im Rückenmark und GCMC

immunhistologisch untersucht. Allerdings fanden sich keine Unterschiede in der

Antigenexpression zwischen BSE-positiven und -negativen Ziegen. Darüber hinaus zeigten

sich keine ultrastrukturellen Synapsenveränderungen im Ganglion cervicale craniale. Ob

Synapsenveränderungen bei Prionenerkrankungen auf das ZNS beschränkt sind, muss jedoch

in weiteren Studien bestätigt werden. Interessanterweise zeigte die vorliegende Studie eine

abnormale Expression des nicht-phosphorylierten Neurofilaments (nNF) in Axonen der

Chapter 6 – Zusammenfassung 59

weißen Substanz des Rückenmarks, welche auf klinisch an BSE erkrankte und PrPSc

-positive

Ziegen beschränkt war. Somit treten wahrscheinlich im Verlauf der BSE-Erkrankung von

Ziegen axonale Schäden und Störungen des axonalen Transports auf. Allerdings muss in

zukünftigen Untersuchungen abgeklärt werden, ob die abnormale Akkumulation von nNF in

einem kausalen Zusammenhang mit gestörten Transportmechanismen im Axon, der

Ausbreitung von Prionen und/oder einer neuronalen Degeneration steht. Des Weiteren wurde

mittels der durchgeführten immunhistologischen Untersuchungen erstmalig eine Ablagerung

von PrPSc

in Gliazellen der weißen Substanz des Rückenmarks bei an BSE erkrankten Ziegen

nachgewiesen, so dass Gliazellen möglicherweise eine wichtige Rolle in der Ausbreitung von

Prionen spielen.

60 Chapter 6 - Zusammenfassung

Chapter 7- References 61

Chapter 7

References

62 Chapter 7 - Reference

Chapter 7: References

ACIN, C., MARTIN-BURRIEL, I., MONLEON, E., LYAHYAI, J., PITARCH, J.L., SERRANO, C.,

MONZÓN, M., ZARAGOZA, P., and BADIOLA, J.J. (2013):

Prion protein gene variability in Spanish goats. inference through susceptibility to classical scrapie strains and

pathogenic distribution of peripheral PrPsc.

PLoS One. 8, e61118.

ACUTIS, P.L., BOSSERS, A., PRIEM, J., RIINA, M.V., PELETTO, S., MAZZA, M., CASALONE, C.,

FORLONI, G., RU, G., and CARAMELLI, M. (2006):

Identification of prion protein gene polymorphisms in goats from Italian scrapie outbreaks.

J. Gen. Virol. 87, 1029-1033.

ACUTIS, P.L., MARTUCCI, F., D’ANGELO, A., PELETTO, S., COLUSSI, S., MAURELLA, C.,

PORCARIO, C., IULINI, B., MAZZA, M., DELL'ATTI, L., ZUCCON, F., CORONA, C.,

MARTINELLI, N., CASALONE, C., CARAMELLI, M., and LOMBARDI, G. (2012):

Resistance to classical scrapie in experimentally challenged goats carrying mutation K222 of the prion protein

gene.

Vet. Res. 43, 8.

AGUILAR-CALVO, P., ESPINOSA, J.C., PINTADO, B., GUTIÉRREZ-ADÁN, A., ALAMILLO, E.,

MIRANDA, A., PRIETO, I., BOSSERS, A., ANDREOLETTI, O., and TORRES, J.M. (2014):

Role of the goat K222-PrPC polymorphic variant in prion infection resistance.

J. Virol. 88, 2670-2676.

AGUZZI, A., and POLYMENIDOU, M. (2004):

Mammalian prion biology: one century of evolving concepts.

Cell 116, 313-327.

AGUZZI, A., BAUMANN, F., and BREMER, J. (2008):

The prion’s elusive reason of being.

Annu. Rev. Neurosci. 341, 439-477.

AGUZZI, A., HEIKENWALDER, M., and POLYMENIDOU, M. (2007):

Insights into prion strains and neurotoxicity.

Nat. Rev. Mol. Cell Biol. 8, 552-561.

ALMER, G., VUKOSAVIC, S., ROMERO, N., and PRZEDBORSKI, S. (1999):

Chapter 7- References 63

Inducible nitric oxide synthase up-regulation in a transgenic mouse model of familial amyotrophic lateral

sclerosis.

J. Neurochem. 72, 2415-2425.

ALPER, T., Haig, D.A., and Clarke, M.C. (1966):

The exceptionally small size of the scrapie agent.

Biochem. Biophys. Res. Commun. 22, 278-284.

ANDRÉOLOTTI, O. (2004):

PrPSc

Immunohistochemistry. In: Methods and tools in biosciences and medicine—techniques in prion research,

Eds. Lehmann S, Grassi J.

Birkhauser Verlag, Berlin.

Pp. 82–96.

ANDRÉOLOTTI, O., BERTHON, P., MARC, D., SARRADIN, P., GROSCLAUDE, J., van KEULEN, L.,

SCHELCHER, F., ELSEN, J.M., and LANTIER, F. (2000):

Early accumulation of PrPSc in gut-associated lymphoid and nervous tissues of susceptible sheep from a

Romanov flock with natural scrapie.

J. Gen. Virol. 81, 3115-3126.

ANONYMOUS, (2001):

Rules for the prevention, control and eradication of certain transmissible spongiform encephalopathies (and

amendments).

European Parliament and the council of the European Union. Regulation (EC) No 999/2001.

BALKEMA-BUSCHMANN, A., FAST, C., KAATZ, M., EIDEN, M., ZIEGLER, U., MCINTYRE, L.,

KELLER, M., HILLS, B., and GROSCHUP, M.H. (2011):

Pathogenesis of classical and atypical BSE in cattle.

Prev. Vet. Med. 102, 112-117.

BARILLET, F., MARIAT, D., AMIGUES, Y., FAUGERAS, R., CAILLAT, H., MOAZAMI-

GOUDARZI, K., RUPP, R., BABILLIOT, J.M., LACROUX, C., LUGAN, S., SCHELCHER, F.,

CHARTIER, C., CORBIÈRE, F., ANDRÉOLETTI, O., and PERRIN-CHAUVINEAU, C. (2009):

Identification of seven haplotypes of the caprine PrP gene at codons 127, 142, 154, 211, 222 and 240 in French

Alpine and Saanen breeds and their association with classical scrapie.

J. Gen. Virol. 90, 769-776.

BARINGER, J.R., BOWMAN, K.A., and PRUSINER, S.B. (1983):

Replication of the scrapie agent in hamster brain precedes neuronal vacuolation.

64 Chapter 7 - Reference

J. Neuropathol. 42, 485-597.

BARON, T. (2002):

Identification of inter-species transmission of prion strains.

J. Neuropathol. Exp. Neurol. 61, 377-383.

BARON, T.M., and BIACABE, A.G. (2001):

Molecular analysis of the abnormal prion protein during coinfection of mice by the bovine spongiform

encephalopathy and a scrapie agent.

J. Virol. 75, 107-114.

BARROW, P.A., HOLMGREN, C.D., TAPPER, A.J., and JEFFERYS, J.G. (1999):

Intrinsic physiological and morphological properties of principal cells of the hippocampus and neocortex in

hamsters infected with scrapie.

Neurobiol. Dis. 6, 406-423.

BEEKES, M., and McBRIDE, P.A. (2007):

The spread of prions through the body in naturally acquired transmissible spongiform encephalopathies.

FEBS J. 274, 588-605.

BEEKES, M., McBRIDE, P.A., and BALDAUF, E. (1998):

Cerebral targeting indicates vagal spread of infection in hamsters fed with scrapie.

J. Gen. Virol. 79, 601-607.

BELL, J.E., and IRONSIDE, J.W. (1993):

Neuropathology of spongiform encephalopathies in humans.

Br. Med. Bull. 49, 738-777.

BELLINGER, K., DIENER, T.O., MCKINLEY, M.P., GROTH, D.F., SMITH, D.R., and PRUSINER,

S.B. (1987):

Purified scrapie prions resist inactivation by procedures that hydrolyze, modify, or shear nucleic acids.

Virol. 160, 271-274.

BELLWORTHY, S.J., HAWKINS, S.A., GREEN, R.B., BLAMIRE, I., DEXTER, G., DEXTER, I.,

LOCKEY, R., JEFFREY, M., RYDER, S., BERTHELIN-BAKER, C., and SIMMONS, M.M. (2005):

Tissue distribution of bovine spongiform encephalopathy infectivity in Romney sheep up to the onset of clinical

disease after oral challenge.

Vet. Rec. 156, 197-202.

Chapter 7- References 65

BENCSIK, A., DEBEER, S., PETIT, T., and BARON, T. (2009):

Possible case of maternal transmission of feline spongiform encephalopathy in a captive cheetah.

PLoS ONE 4, 6929.

BERINGUE, V., MALLINSON, G., KAISAR, M., TAYEBI, M., SATTAR, Z., JACKSON, G., ANSTEE,

D., COLLINGE, J., and HAWKE, S. (2003):

Regional heterogeneity of cellular prion protein isoforms in the mouse brain.

Brain 126, 2065-2073.

BERINGUE, V., VILOTTE, J.L., and LAUDE, H. (2008):

Review article: Prion agent diversity and species barrier.

Vet. Res. 39, 47.

BESSEN, R.A., and MARSH, R.F. (1992):

Biochemical and physical properties of the prion protein from two strains of the transmissible mink

encephalopathy agent.

J. Virol. 66, 2096-2101.

BESSEN, R.A., and MARSH, R.F. (1994):

Distinct PrP properties suggest the molecular basis of strain variation in transmissible mink encephalopathy.

J. Virol. 68, 7859-7868.

BOLANOS, J.P., ALMEIDA, A., FERNANDEZ, E., MEDINA, J.M., LAND, J.M., CLARK, J.B., and

HEALES, S.J. (1997):

Potential mechanisms for nitric oxide-mediated impairment of brain mitochondrial energy metabolism.

Biochem. Soc. Trans. 25, 944-949.

BOLTON, D.C. (1998):

Prion distribution in hamster lung and brain following intraperitoneal inoculation.

J. Gen. Virol. 79, 2557-2562.

BOLTON, D.C., RUDELLI, R.D., CURRIE, J.R., and BENDHEIM, P.E. (1991):

Copurification of Sp33-37 and scrapie agent from hamster brain prior to detectable histopathology and clinical

disease.

J. Gen. Virol. 72, 2905-2913.

BOUZALAS, I.G., DOVAS, C.I., BANOS, G., PAPANASTASOPOULOU, M., KRITAS, S.,

OEVERMANN, A., PAPAKOSTAKI, D., EVANGELIA, C., PAPADOPOULOS, O., SEUBERLICH, T.,

and KOPTOPOULOS, G. (2010):

66 Chapter 7 - Reference

Caprine PRNP polymorphisms at codons 171, 211, 222 and 240 in a Greek herd and their association with

classical scrapie.

J. Gen. Virol. 91, 1629-1634.

BROWN, D.R. (2001):

Prion and prejudice: normal protein and the synapse.

Trends Neurosci. 24, 85-90.

BROWN, P., and CERVENAKOVA, L. (2005):

A prion lexicon (out of control).

Lancet 365, 122.

BROWN, P., and GAJDUSEK, D.C. (1991):

Survival of scrapie virus after 3 years' interment.

Lancet 337, 269-270.

BRUCE, M. (1996):

Strain typing studies of scrapie and BSE. In methods in molecular medicine: Prion diseases (Baker, H. et al.,

Eds.),

Pp. 223-236.

BRUCE, M., CHREE, A., MCCONNELL, I., FOSTER, J., PEARSON, G., and FRASER, H. (1994):

Transmission of bovine spongiform encephalopathy and scrapie to mice: strain variation and the species barrier.

Philos. Trans. R. Soc. Lond. B. Biol. Sci. 343, 405-411.

BRUCE, M.E. (1993):

Scrapie strain variation and mutation.

Br. Med. Bull. 49, 822-838.

BRUCE, M.E., and DICKINSON, A.G. (1987):

Biological evidence that scrapie agent has an independent genome.

J. Gen. Virol. 68, 79-89.

BRUCE, M.E., BOYLE, A., COUSENS, S., MCCONNELL, I., FOSTER, J., GOLDMANN, W., and

FRASER, H. (2002):

Strain characterization of natural sheep scrapie and comparison with BSE.

J. Gen. Virol. 83, 695-704.

Chapter 7- References 67

BRUCE, M.E., FRASER, H., MCBRIDE, P.A., SCOTT, J.R., and DICKINSON, A.G. (1992):

The basis of strain variation in scrapie, in prion diseases of humans and animals (PRUSINER, S. B.,

COLLINGE, J., POWELL, J., AND ANDERTON, B., edS.), Ellis Horwood, Chichester,

Pp. 497-508.

BUDKA, H. (2003):

Neuropathology of prion diseases.

Br. Med. Bull. 66, 121-130.

BUDKA, H., AGUZZI, A., BROWN, P., BRUCHER, J.M., BUGIANI, O., GULLOTTA, F., HALTIA, M.,

HAUW, J.J., IRONSIDE, J.W., and JELLINGER, K. (1995):

Neuropathological diagnostic criteria for Creutzfeldt-Jakob disease (CJD) and other human spongiform

encephalopathies.

Brain Pathol., 4, 459-466.

BUELER, H., AGUZZI, A., SAILER, A., GREINER,R.A., AUTENRIED, P., AGUET, M., and

WEISSMANN, C. (1993):

Mice devoid of PrP are resistant to scrapie.

Cell 73, 1339-1347.

BUELER, H., FISCHER, M., LANG, Y., BLUETHMANN, H., LIPP, H.P., DEARMOND, S.J.,

PRUSINER, S.B., AGUET, M., and WEISSMANN, C. (1992):

Normal development and behaviour of mice lacking the neuronal cell surface PrP protein. Nature 356, 577-582.

BUSCHMANN, A., and GROSCHUP, M.H. (2005):

Highly bovine spongiform encephalopathy sensitive transgenic mice confirm the essential restriction of

infectivity to the nervous system in clinically diseased cattle.

J. Infect. Dis. 192, 934-942.

BUSCHMANN, A., PFAFF, E., REIFENBERG, K., MÜLLER, H.M., and GROSCHUP, M.H. (2000):

Detection of cattle-derived BSE prions using transgenic mice overexpressing bovine PrPC.

Arch. Virol. Suppl. 16, 75-86.

CARLSON, G.A., GOODMAN, P.A., LOVETT, M., BENJAMIN, A., TAYLOR, B.A., MARSHALL,

S.T., PETERSON-TORCHIA, M., WESTAWAY, D., and PRUSINER, S.B. (1988):

Genetics and polymorphism of the mouse prion gene complex: control of scrapie incubation time.

Mol. Cell. Biol. 8, 5528-5540.

68 Chapter 7 - Reference

CASTELLANI, R., HIRAI, K., ALIEV, G., DREW, K.L., NUNOMURA, A., TAKEDA, A., CASH, A.D.,

OBRENOVICH, M.E., PERRY, G., and SMITH, M.A, (2002):

Role of mitochondrial dysfunction in Alzheimer’s disease.

J. Neurosci. Res. 70, 357-360.

CAUGHEY, B., and RAYMON, G. (1991):

The scrapie associated form of PrP is made from a cell surface precursor that is both protease and

phospholipase-sensitive.

J. Biol. Chem. 266, 18217-18223.

CHANDLER, R.L. (1961):

Encephalopathy in mice produced by inoculation with scrapie brain material.

Lancet 1, 1378-1379.

CHANDLER, R.L. (1971):

Intramammary inoculation of mice with scrapie.

Br. Vet. J. 127, 1-2.

CHANDLER, R.L., and FISHER, J. (1963):

Experimental transmission of scrapie to rats.

Lancet 2, 1165.

CHANDLER, R.L., and TURFREY, B.A. (1972):

Inoculation of voles, Chinese hamsters, gerbils and guinea-pigs with scrapie brain material.

Res. Vet. Sci. 13, 219-224.

CHESEBRO, B. (2003):

Introduction to the transmissible spongiform encephalopathies or prion disease.

Br. Med. Bull. 66, 1-20.

CHESEBRO, B., TRIFILO, M., RACE, R., MEADE-WHITE, K., TENG, C., LACASSE, R.,

RAYMOND, L., FAVARA, C., BARON, G., PRIOLA, S., CAUGHEY, B., MASLIAH, E., and

OLDSTONE, M. (2005):

Anchorless prion protein results in infectious amyloid disease without clinical scrapie.

Science 308, 1435-1439.

CLEETER, M.W., COOPER, J.M., DARLEY-USMAR, V.M., MONCADA, S., and SCHAPIRA, A.H.

(1994):

Chapter 7- References 69

Reversible inhibition of cytochrome c oxidase, the terminal enzyme of the mitochondrial respiratory chain, by

nitric oxide. Implications for neurodegenerative diseases.

FEBS Lett. 345, 50-54.

CLINTON, J., FORSYTH, C., ROYSTON, M.C., and ROBERTS, G.W. (1993):

Synaptic degeneration is the primary neuropathological feature in prion disease: a preliminary study.

Neuroreport. 4, 65-68.

COLLINGE, J. (2001):

Prion diseases of humans and animals: Their Causes and Molecular Basis.

Annu. Rev. Neurosci. 24, 519-550.

COLLINGE, J., BECK, J., CAMPBELL, T., ESTIBEIRO, K., and WILL, R.G. (1996):

Prion protein gene analysis in new variant cases of Creutzfeldt-Jakob disease.

Lancet 348, 56.

COLLINGE, J., WHITTINGTON, M.A., SIDLE, K.C., SMITH, C.J., PALMER, M.S., CLARKE, A.R.,

and JEFFERYS, J.G. R. (1994):

Prion protein is necessary for normal synaptic function.

Nature 370, 295-297.

CORBIÈRE, F., PERRIN-CHAUVINEAU, C., LACROUX, C., COSTES, P., THOMAS, M., BRÉMAUD,

I., MARTIN, S., LUGAN, S., CHARTIER, C., SCHELCHER, F., BARILLET, F., and ANDREOLETTI,

O. (2013):

PrP-associated resistance to scrapie in five highly infected goat herds.

J. Gen. Virol. 94, 241-245.

CRONIER, S., LAUDE, H., and PEYRIN, J.M. (2004):

Prions can infect primary cultured neurons and astrocytes and promote neuronal cell death.

Proc. Natl. Acad. Sci. USA 101, 12271-12276.

CROZET, C., BENCSIK, A., FLAMANT, F., LEZMI, S., SAMARUT, J., and BARON, T. (2001b):

Florid plaques in ovine PrP transgenic mice infected with an experimental ovine BSE.

EMBO Rep. 2, 952-956.

CROZET, C., FLAMANT, F., BENCSIK, A., AUBERT, D., SAMARUT, J., and BARON, T. (2001a):

Efficient transmission of two different sheep scrapie isolates in transgenic mice expressing the ovine PrP gene.

J. Virol. 75, 5328-5334.

70 Chapter 7 - Reference

CHELLE, P.L. (1942):

Un cas de tremblante chez la chevre.

Bull Acad. Vet. Fr. 15, 294-295.

CUILLE, J., and CHELLE, P.L. (1936):

Pathologie animale – La maladie dite tremblante du mouton est-elle inoculable ?

Comptes rendus hebdomadaires des seances de l’Academie des Sciences 203, 1552-1554.

CUILLE, J., and CHELLE, P.L. (1939):

Transmission experimentale de la tremblante a la chevre.

Comptes rendus hebdomadaires des siences de l’Academie des Sciences 208, 1058-1060.

CUNNINGHAM, C., DEACON, R., WELLS, H., BOCHE, D., WATERS, S., DINIZ, C.P., SCOTT, H.,

RAWLINS, J.N., and PERRY, V.H. (2003):

Synaptic changes characterize early behavioural signs in the ME7 model of murine prion disease.

Eur. J. Neurosci. 17, 2147-2155.

DeMARCO, M.L., and DAGGETT, V. (2004):

From conversion to aggregation: protofibril formation of the prion protein.

Proc. Natl. Acad. Sci. U.S.A. 101, 2293-2298.

DETWILER, L.A. (1992):

Scrapie.

Rev. Sci. Tech. 11, 491-537.

DETWILER, L.A., and BAYLIS, M. (2003):

The epidemiology of scrapie.

Rev. Sci. Tech. 22, 121-143.

DIEDRICH, J.F., BENDHEIM, P.E., KIM, Y.S., CARP, R.I., and HAASE, A.T. (1991):

Scrapie-associated prion protein accumulates in astrocytes during scrapie infection.

Proc. Natl. Acad. Sci. USA 88, 375-379.

ERMOLAYEV, V., CATHOMEN, T., MERK, J., FRIEDRICH, M., HÄRTIG, W., HARMS, G.S.,

KLEIN, M.A., and FLECHSIG, E. (2009):

Impaired axonal transport in motor neurons correlates with clinical prion disease.

Plos Pathog. 5, e1000558.

Chapter 7- References 71

FERNÁNDEZ-CHACÓN, R., WÖLFEL, M., NISHIMUNE, H., TABARES, L., SCHMITZ, F.,

CASTELLANO-MUÑOZ, M., ROSENMUND, C., MONTESINOS, M.L., SANES, J.R.,

SCHNEGGENBURGER, R., and SÜDHOF, T.C. (2004):

The synaptic vesicle protein CSP alpha prevents presynaptic degeneration.

Neuron 42, 237-251.

FERRER, I. (2002):

Synaptic pathology and cell death in the cerebellum in Creutzfeldt-Jakob disease.

Cerebellum 1, 213-222.

FERRER, I., PUIG, B., BLANCO, R., and MARTI, E. (2000):

Prion protein deposition and abnormal synaptic protein expression in the cerebellum in Creutzfeldt-Jakob

disease.

Neuroscience 97, 715-726.

FERRER, I., RIVERA, R., BLANCO, R., and MART, E. (1999):

Expression of proteins linked to exocytosis and neurotransmission in patients with Creutzfeldt-Jakob disease.

Neurobiol. Dis. 6, 92-100.

FISCHER, M., RULICKE1,T., RAEBER, A., SAILER, A., MOSER, M., OESCH, B., BRANDNER, S.,

AGUZZI, A., and WEISSMANN, C. (1996):

Prion protein (PrP) with amino-proximal deletions restoring susceptibility of PrP knockout mice to scrapie.

EMBO J. 15, 1255-1264.

FOSTER, J.D., BRUCE, M., McCONNELL, I., CHREE, A., and FRASER, H. (1996):

Detection of BSE infectivity in brain and spleen of experimentally infected sheep.

Vet. Rec. 138, 546-548.

FOSTER, J.D., PARNHAM, D., CHONG, A., GOLDMANN, W., and HUNTER, N. (2001a):

Clinical signs, histopathology and genetics of experimental transmission of BSE and natural scrapie to sheep

and goats.

Vet. Rec. 148, 165-171.

FOSTER, J.D., PARNHAM, D.W., HUNTER, N., and BRUCE, M. (2001b):

Distribution of the prion protein in sheep terminally affected with BSE following experimental oral

transmission.

J. Gen. Virol. 82, 2319-2326.

FOURNIER, J.G., ESCAIG-HAYE, F., BILLETTE DE VILLEMEUR, T., and ROBAIN, O. (1995):

72 Chapter 7 - Reference

Ultrastructural localization of cellular prion protein (PrPC) in synaptic boutons of normal hamster hippocampus.

CR Acad. Sci. III 318, 339-344.

FRAGKIADAKI, E.G., VACCARI, G., EKATERINIADOU, L.V., AGRIMI, U., GIADINIS, N.D.,

CHIAPPINI, B., ESPOSITO, E., CONTE, M., and NONNO, R. (2011):

PRNP genetic variability and molecular typing of natural goat scrapie isolates in a high number of infected

flocks.

Vet. Res. 42, 104.

FRASER, H., and DICKINSON, A.G. (1973):

Scrapie in mice: agent-strain differences in the distribution and intensity of grey matter vacuolation.

J. Comp. Pathol. 83, 29-40.

FRASER, H., BRUCE, M.E., CHREE, A., MCCONNELL, I., and WELLS, G.A.H. (1992):

Transmission of bovine spongiform encephalopathy and scrapie to mice.

J. Gen. Virol. 73, 1891-1897.

FRASER, J.R. (2002):

What is the basis of transmissible spongiform encephalopathy induced neurodegeneration and can it be

repaired?

Neuropathol. Appl. Neurobiol. 28, 1-11.

FUKUDA, S., ONOE, S., NIKAIDO, S., FUJII, K., KAGEYAMA, S., IWAMARU, Y., IMAMURA, M.,

MASUJIN, K., MATSUURA, Y., SHIMIZU, Y., KASAI, K., YOSHIOKA, M., MURAYAMA, Y.,

MOHRI, S., YOKOYAMA, T., and OKADA, H. (2012):

Neuroanatomical distribution of disease-associated prion protein in experimental bovine spongiform

encephalopathy in cattle after intracerebral inoculation.

Jpn. J. Infect. Dis. 65, 37-44.

GABIZON, R., TELLING, G., MEINER, Z., HALIMI, M., KAHANA, I., and PRUSINER, S.B. (1996):

Insoluble wild-type and protease-resistant mutant prion protein in brains of patients with inherited prion

diseases.

Nat. Med. 2, 59-64.

GAJDUSEK, D.C. (1977):

Unconventional viruses and the origin and disappearance of kuru.

Science 197, 943-960.

GAJDUSEK, D.C. (1985):

Chapter 7- References 73

Hypothesis: interference with axonal transport of neurofilaments as a common pathogenetic mechanism in

certain diseases of the central nervous system.

N. Engl. J. Med. 312, 714-719.

GAJDUSEK, D.C., and ZIGAS, V. (1957):

Degenerative disease of the central nervous system in New Guinea; the endemic occurrence of kuru in the native

population.

N. Engl. J. Med. 257, 974-978.

GAMBETTI, P., PETERSEN, R.B., PARCHI, P., CHEN, S.G., CAPELLARI, S., and GOLDFARB, L.

(2004):

Inherited prion diseases. In: Prusiner SB, eds. Prion Biology and Diseases.

Cold Spring Harbor Laboratory

Pp. 673-775.

GAVIER-WIDEN, D., STACK, M. J., BARON, T., BALACHANDRAN, A., and SIMMONS, M. (2005):

Diagnosis of transmissible spongiform encephalopathies in animals: a review.

J. Vet. Diagn. Invest. 17, 509-527.

GIBBS, C. J., ASHER, D.M., KOBRINE, A., AMYX, H.L., SULIMA, M.P., and GAJDUSEK, D.C.

(1994):

Transmission of Creutzfeldt-Jakob disease to a chimpanzee by electrodes contaminated during neurosurgery.

J. Neurol. Neurosurg. Psych. 57, 757-758.

GONZÁLEZ, L., MARTIN, S., and JEFFREY, M. (2003):

Distinct profiles of PrP(d) immunoreactivity in the brain of scrapie and BSE-infected sheep: implications for

differential cell targeting and PrP processing. J Gen Virol 84: 1339–1350.

GONZÁLEZ, L., MARTIN, S., BEGARA-MCGORUM, I., HUNTER, N., HOUSTON, F., SIMMONS,

M., and JEFFREY, M. (2002):

Effects of agent strain and host genotype on PrP accumulation in the brain of sheep naturally and experimentally

affected with scrapie.

J. Comp. Pathol. 126, 17-29.

GONZÁLEZ, L., MARTIN, S., HOUSTON, F.E., HUNTER, N., REID, H.W., BELLWORTHY, S.J., and

JEFFREY, M. (2005):

Phenotype of disease-associated PrP accumulation in the brain of bovine spongiform encephalopathy

experimentally infected sheep.

J. Gen. Virol. 86, 827-838.

74 Chapter 7 - Reference

GOODFIELD, J. (1997)

Cannibalism and kuru.

Nature 387, 841.

GRAY, B.C., SISKOVA, Z., PERRY, V.H., and O’CONNOR, V. (2009):

Selective presynaptic degeneration in the synaptopathy associated with ME7-induced hippocampal pathology.

Neurobiol. Dis. 35, 63-74.

GRIFFITH, J.S. (1967):

Self replication and scrapie.

Nature 215, 1043-1044.

GROSCHUP, M.H., and BUSCHMANN, A. (2008):

Rodent models for prion diseases.

Vet. Res. 39, 32.

HAEBERLE, A.M., RIBAUT-BARASSIN, C., BOMBARDE, G., MARIANI. J., HUSSMANN, G.,

GRASSI, J., and BAILLY, Y. (2000):

Synaptic prion immunoreactivity in the rodent cerebellum.

Microsc. Res. Tech. 50, 66-76.

HAÏK, S., and BRANDEL, J.P. (2014):

Infectious prion diseases in humans: cannibalism, iatrogenicity and zoonoses.

Infect. Genet. Evol. 26, 303-312.

HE, L., LU, X.Y., JOLLY, A.F., ELDRIDGE, A.G.,WATSON, S.J., JACKSON, P.K., BARSH, G.S., and

GUNN, T.M. (2003):

Spongiform degeneration in mahoganoid mutant mice.

Science 299, 710-712.

HILL, A., DESBRUSLAIS, M., JOINER, S., KATIE, C., SIDLE, L., GOWLAND, I., COLLINGE, J.,

DOEY, L. J., and LANTOS, P. (1997):

The same prion strain causes vCJD and BSE.

Nature 389, 448-450.

HILTON, K.J., CUNNINGHAM, C., REYNOLDS, R.A., and PERRY, V.H. (2013):

Early Hippocampal Synaptic Loss Precedes Neuronal Loss and Associates with Early Behavioural Deficits in

Three Distinct Strains of Prion Disease.

PLoS One. 8, e68062.

Chapter 7- References 75

HOLLISTER, J.R., LEE, K.S., DORWARD, D.W., and BARON, G.S. (2015):

Efficient Uptake and Dissemination of Scrapie Prion Protein by Astrocytes and Fibroblasts from Adult Hamster

Brain.

PLoS One 10, e0115351.

HORNEMANN, S., KORTH, C., OESCH, B., RIEK, R., WIDER, G., WÜTHRICH, K., and

GLOCKSHUBER, R. (1997):

Recombinant full-length murineprion protein, mPrP(23–231): purification and spectroscopic characterization.

FEBS Lett. 413, 277-281.

IULINI, B., MAURELLA, C., PINTORE, M.D., VALLINO, C.E., CORBELLINI, D., PORCARIO, C.,

PAUTASSO, A., SALATA, C., GELMETTI, D., AVANZATO, T., PALÙ, G., D'ANGELO, A.,

CARAMELLI, M., CASALONE, C. (2012):

Ten years of BSE surveillance in Italy: neuropathological findings in clinically suspected cases.

Res. Vet. Sci. 93, 872-878.

JEFFREY, M., and GONZÁLEZ, L. (2004):

Pathology and pathogenesis of bovine spongiform encephalopathy and scrapie.

Curr. Topics Microbiol. Immunol. 284, 65-97.

JEFFREY, M., and GONZÁLEZ, L. (2007):

Classical sheep transmissible spongiform encephalopathies: pathogenesis, pathological phenotypes and clinical

disease.

Neuropath. Appl. Neurobiol. 33, 373-394.

JEFFREY, M., HALLIDAY, W.G., BELL, J., JOHNSTON, A.R., MACLEOD, N.K., INGHAM, C.,

SAYERS, A.R., BROWN, D.A., and FRASER, J.R. (2000):

Synapse loss associated with abnormal PrP precedes neuronal degeneration in the scrapie-infected murine

hippocampus.

Neuropathol. Appl. Neurobiol. 26, 41-54.

KAATZ, M., FAST, C., ZIEGLER, U., BALKEMA-BUSCHMANN, A., HAMMERSCHMIDT, B.,

KELLER, M., OELSCHLEGEL, A., McINTYRE, L., and GROSCHUP, M. (2012):

Spread of classical BSE prions from the gut via the peripheral nervous system to the Brain.

Immunopathol. Infect. Dis. 181, 515-524.

KIMBERLIN, R.H., and WALKER, C.A. (1977):

Characteristics of a short incubation model of scrapie in the golden hamster.

J. Gen. Virol. 34, 295-304.

76 Chapter 7 - Reference

KIMBERLIN, R.H., and WALKER, C.A. (1978):

Evidence that the transmission of one source of scrapie agent to hamsters involves separation of agent strains

from a mixture.

J. Gen. Virol. 39, 487-496.

KIMBERLIN, R.H., and WALKER, C.A. (1989):

Pathogenesis of scrapie in mice after intragastric infection.

Virus Res. 12, 213-220.

KIMBERLIN, R.H., COLE, S., and WALKER, C.A. (1987):

Temporary and permanent modifications to a single strain of mouse scrapie on transmission to rats and

hamsters.

J. Gen. Virol. 68, 1875-1881.

KITAMOTO, T., SHIN, R.W., DOH-URA, K., TOMOKANE, N., MIYAZONO, M., MURAMOTO, T.,

and TATEISHI, J. (1992):

Abnormal isoform of prion proteins accumulates in the synaptic structures of the central nervous system in

patients with Creutzfeldt-Jakob disease.

Am. J. Pathol. 140, 1285-1294.

KLEIN, M.A., FRIGG, R., FLECHSIG, E., RAEBER, A.J., KALINKE, U., BLUETHMANN, H.,

BOOTZ, F., SUTER, M., ZINKERNAGEL, R.M., and AGUZZI, A. (1997):

A crucial role for B cells in neuroinvasive scrapie.

Nature 390, 687-690.

KONOLD, T., GOLDMANN, W., CAWTHRAW, S., HAWKINS, A.C., GEMMA., E.B., PHELAN, L.J.,

SIMMONS, M.M., GONZÁLEZ, L., and SISÓ, S. (2010):

Monitoring of clinical signs in goats with transmissible spongiform encephalopathies.

BMC Vet. Res. 6, 13.

KRETZSCHMAR, H.A. (1993):

Human prion diseases (spongiform encephalopathies).

Arch. Virol. Suppl. 7, 261-293.

KUJALA, P., RAYMOND, C.R., ROMEIJN, M., GODSAVE, S.F., van KASTEREN, S.I., WILLE, H.,

PRUSINER, S.B., MABBOTT, N.A., and PETERS, P.J. (2011):

Prion uptake in the gut: identification of the first uptake and replication sites.

PLoS Pathog. 7, e1002449.

Chapter 7- References 77

KÜNZI, V., GLATZEL, M., NAKANO, M.Y., GREBER, U.F., LEUVEN, F.V., and AGUZZI, A. (2002):

Unhampered Prion Neuroinvasion despite Impaired Fast Axonal Transport in Transgenic Mice Overexpressing

Four Repeat Tau.

J. Neurosci. 22, 7471-7477.

LACROUX, C., PERRIN-CHAUVINEAU, C., CORBIÈRE, F., ARON, N., AGUILAR-CALVO, P.,

TORRES, J.M., COSTES, P., BRÉMAUD, I., LUGAN, S., SCHELCHER, F., BARILLET, F., and

ANDRÉOLETTI, O. (2014):

Genetic resistance to scrapie infection in experimentally challenged goats.

J. Virol. 88, 2406-2413.

LASMÉZAS, C. I., COMOY, E., HAWKINS, S., HERZOG, C., MOUTHON, F., KONOLD, T., AUVRÉ,

F., CORREIA, E., LESCOUTRA-ETCHEGARAY, N., SALÈS, N., WELLS, G., BROWN, P., and

DESLYS, J.P. (2005):

Risk of oral infection with bovine spongiform encephalopathy agent in primates.

Lancet 365, 781-783.

LEE, J., KIM, S.Y., HWANG, K.J., JU, Y.R., and WOO, H.J. (2013):

Prion diseases as transmissible zoonotic diseases.

Osong Public Health Res. Perspect. 4, 57-66.

LEZMI, S., SEUBERLICH, T., OEVERMANN, A., BARON, T., and BENCSIK, A. (2011):

Comparison of brain PrPd distribution in ovine BSE and scrapie.

Vet. Pathol. 48, 1101-1108.

LI, Z., OKAMOTO, K., HAYASHI, Y., and SHENG, M. (2004):

The importance of dendritic mitochondria in the morphogenesis and plasticity of spines and synapses.

Cell 119, 873-887.

LIBERSKI, P.P. (2012):

Historical overview of prion diseases: a view from afar.

Folia Neuropathol. 50, 1-12.

LIBERSKI, P.P., and BUDKA, H. (1999):

Neuroaxonal pathology in Creutzfeldt-Jakob disease.

Acta Neuropathol. 97, 329-334.

LIBERSKI, P.P., KORDEK, R., BROWN, P., and GAJDUSEK, D.C. (1998):

78 Chapter 7 - Reference

Astrocytes in transmissible spongiform encephalopathies (prion diseases), in: Astrocytes in Brain Aging and

Neurodegeneration, edited by H. M. Schipper (R. G. Landes Company, Austin, Texas, USA)

pp. 127–164.

LLOYD, S.E., ONWUAZOR, O.N., BECK, J.A., MALLINSON, G., FARRALL, M., TARGONSKI, P.,

COLLINGE, J., and ELIZABETH, M.C.F. (2001):

Identification of multiple quantitative trait loci linked to prion disease incubation period in mice.

Proc. Natl. Acad. Sci. USA 98, 6279-6283.

MALLUCCI, G., DICKINSON, A., LINEHAN, J., KLOHN, P.C., BRANDNER, S., and COLLINGE, J.

(2003):

Depleting neuronal PrP in prion infection prevents disease and reverses spongiosis.

Science 302, 871-874.

MANSON, J.C., JAMIESON, E., BAYBUTT, H., TUZI, N.L., BARRON, R., MCCONNELL, I.,

SOMERVILLE, R., IRONSIDE, J., WILL, R., SY, M.S., MELTON, D.W., HOPE, J., and BOSTOCK, C.

(1999):

A single amino acid alteration (101L) introduced into murine PrP dramatically alters incubation time of

transmissible spongiform encephalopathy.

EMBO J. 18, 6855-6864.

MARSH, R.F., and HADLOW, W.J. (1992):

Transmissible mink encephalopathy.

Rev. Sci. Tech. 11, 539-550.

MASTRIANNI, J.A., NIXON, R., LAYZER, R., TELLING, G.C., HAN, D., DEARMOND, S.J., and

PRUSINER, S.B. (1999):

Prion protein conformation in a patient with sporadic fatal insomnia.

NEJM 340, 1630-1638.

MATHIASON, C. K., POWERS, J.G., DAHMES. J.S, OSBORN, D.A., MILLER, K.V., WARREN, R.J.,

MASON, G.L. HAYS, S.A., HAYES-KLUG, J., SEELIG, D.M., WILD, M.A., WOLFE, L.L., SPRAKER,

T.R., MILLER, M.W., SIGURDSON, C.J., TELLING, G.C., and HOOVER., E.A. (2006):

Infectious Prions in the Saliva and Blood of Deer with Chronic Wasting Disease.

Science 314, 133-136.

McBRIDE, P.A., SCHULZ-SCHAEFFER, W.J., DONALDSON, M., BRUCE, M., DIRINGER, H.,

KRETZSCHMAR, H.A., and BEEKES, M. (2001):

Early spread of scrapie from the gastrointestinal tract to the central nervous system involves autonomic fibers of

Chapter 7- References 79

the splanchnic and vagus nerves.

J. Virol. 75, 9320-9327.

McGOVERN, G., MARTIN, S., JEFFREY, M., BELLWORTHY, S.J., SPIROPOULOS, J., GREEN, R.,

LOCKEY. R., VICKERY, C.M., THURSTON, L., DEXTER, G., HAWKINS, S.A., and GONZÁLEZ, L.

(2015):

Influence of Breed and Genotype on the Onset and Distribution of Infectivity and Disease-associated Prion

Protein in Sheep Following Oral Infection with the Bovine Spongiform Encephalopathy Agent.

J. Comp. Pathol. 152, 28-40.

McKINLEY, M.P., BOLTON, D.C., and PRUSINER S.B. (1983):

A protease resistant protein is a structural component of the scrapie prion.

Cell 35, 57-62.

MEIER, P., GENOUD, N., PRINZ, M., MAISSEN, M., RÜLICKE, T., ZURBRIGGEN, A., RAEBER,

A.J., and AGUZZI, A. (2003):

Soluble dimeric prion protein binds PrP(Sc) in vivo and antagonizes prion disease.

Cell 113, 49-60.

MILLER, C.C., ACKERLEY, S., BROWNLEES, J., GRIERSON, A.J., JACOBSEN, N.J., and

THORNHILL, P. (2002):

Axonal transport of neurofilaments in normal and disease states.

Cell Mol. Life Sci. 59, 323-330.

MORINET, F. (2014):

Prions: a model of conformational disease?

Pathol. Biol. 62, 96-99.

MURAMOTO, T., DEARMOND, S.J., SCOTT, M., TELLING, G.C., COHEN, F.E., and PRUSINER,

S.B. (1997):

Heritable disorder resembling neuronal storage disease in mice expressing prion protein with deletion of an

alpha-helix.

Nat. Med. 3, 750-755.

PAN, K.M., (1993):

Conversion of alpha-helices into beta-sheets features in the formation of the scrapie prion proteins.

Proc. Natl. Acad. Sci. USA. 90, 10962-10966.

80 Chapter 7 - Reference

PAPASAVVA-STYLIANOU, P., WINDL, O., SAUNDERS, G., MAVRIKIOU, P., TOUMAZOS, P., and

KAKOYIANNIS, C. (2011):

PrP gene polymorphisms in Cyprus goats and their association with resistance or susceptibility to natural

scrapie.

Vet. J. 187, 245-250.

PATTISON, I.H., and JONES, K.M. (1967):

The possible nature of the transmissible agent of scrapie.

Vet. Rec. 80, 2-9.

PATTISON, I.H., GORDON, W.S., and MILLSON, G.C. (1959):

Experimental production of scrapie in goats.

J. Comp. Pathol. 69, 300-312.

PICANCO-DINIZ, C.W., BOCHE, D., GOMES-LEAL, W., PERRY, V.H., and CUNNINGHAM, C.

(2004):

Neuropil and neuronal changes in hippocampal NADPH-diaphorase histochemistry in the ME7 model of murine

prion disease.

Neuropathol. Appl. Neurobiol. 30, 292-303.

POST, K., RIESNER, D., WALLDORF, V., and MEHLHORN, H. (1999):

Fly larvae and pupae as vectors for scrapie.

Lancet. 354, 1969-1970.

PRITZKOW, S., WAGENFUHR, K., DAUS, M.L., BOERNER, S., LEMMER, K., THOMZIG, A.,

MIELKE, M., and BEEKES, M. (2011):

Quantitative detection and biological propagation of scrapie seeding activity in vitro facilitate use of prions as

model pathogens for disinfection.

PLoS ONE 6, e20384.

PRUSINER, S.B., GROTH, D.F., COCHRAN, S.P., MASIARZ, F.R., MCKINLEY, M.P., and

MARTINEZ, H.M. (1980):

Molecular properties, partial purification, and assay by incubation period measurements of the hamster scrapie

agent.

Biochem. 19, 4883-4891.

PRUSINER, S.B. (1982):

Novel proteinaceous infectious particles cause scrapie.

Science 216, 136-144.

Chapter 7- References 81

PRUSINER, S.B. (1998):

Prions.

Proc. Natl. Acad. Sci. USA 95, 13363-13383.

PRUSINER, S.B. (2013):

Biology and genetics of prions causing neurodegeneration.

Annu. Rev. Genet. 47, 601-623.

PRUSINER, S.B., GROTH, D., SERBAN, A., KOEHLER, R., FOSTER, D., TORCHIA, M., BURTON,

D., YANG, S-L., and DEARMOND, S.J. (1993):

Ablation of the prion protein (PrP) gene in mice prevents scrapie and facilitates production of anti-PrP

antibodies.

Proc. Natl. Acad. Sci. 90, 10608-10612.

PRUSINER, S.B., SCOTT, M., FOSTER, D., PAN, K-M., GROTH, D., MIRENDA, TORCHIA, C., M.,

YANG S-L., SERBAN, D., CARLSON, G.A., HOPPE, P.C., WESTAWAY, D., and DEARMOND S.J.

(1990):

Transgenetic studies implicate interactions between homologous PrP isoforms in scrapie prion replication.

Cell 63, 673-686.

RACE, R., and CHESEBRO, B. (1998):

Scrapie infectivity found in resistant species.

Nature 392, 770.

RACE, R., JENNY, A., and SUTTON, D. (1998):

Scrapie infectivity and proteinase K-resistant prion protein in sheep placenta, brain, spleen, and lymph node:

implications for transmission and antemortem diagnosis.

J. Infect. Dis. 178, 949-953.

RAEBER, A.J., RACE, R.E., BRANDNER, S., PRIOLA, S.A., SAILER, A., BESSEN, R.A., MUCKE, L.,

MANSON, J., AGUZZI, A., OLDSTONE, M.B., WEISSMANN, C., and CHESEBRO, B. (1997):

Astrocyte-specific expression of hamster prion protein (PrP) renders PrP knockout mice susceptible to hamster

scrapie.

EMBO J. 16, 6057-6065.

RAVILOCHAN, K., and TYLER, K.L. (1992):

Human transmissible neurodegenerative diseases (prion diseases).

Semin. Neurol. 12, 178-192.

82 Chapter 7 - Reference

REIS, R., HENNESSY, E., MURRAY, C., GRIFFIN, É.W., and CUNNINGHAM, C. (2015):

At the centre of neuronal, synaptic and axonal pathology in murine prion disease: degeneration of

neuroanatomically linked thalamic and brainstem nuclei.

Neuropathol. Appl. Neurobiol. doi. 10.1111/nan.12232.

RICHARD, J., WHITLEY, M.D., NONI MACDONALD, M.D., DAVID, M., and ASHER, M.D. (2000):

Technical report: transmissible spongiform encephalopathies: A Review for pediatricians.

Pediatrics. 106, 160-165.

RIESNER, D., (2003):

Biochemistry and structure of PrP(C)

and PrP(Sc)

.

Br. Med. Bull. 66, 21-33.

RIEK, R., HORNEMANN, S., WIDER, G., BILLETER, M., GLOCKSHUBER, R., and WUTHRICH, K.

(1996):

NMR structure of the mouse prion protein domain PrP(121–321).

Nature 382, 180-182.

RUSSELAKIS-CARNEIRO, M., HETZ, C., MAUNDRELL, K., and SOTO, C. (2004):

Prion replication alters the distribution of synaptophysin and caveolin 1 in neuronal lipid rafts.

Am. J. Pathol. 165, 1839-1848.

SAFAR, J., WILLE, H., ITRRI, V., GROTH, D., SERBAN, H., TORCHIA, M., COHEN, F.E., and

PRUSINER, S.B. (1998):

Eight prion strains have PrPSc

molecules with different conformations.

Nat. Med. 4, 1157-1165.

SAILER, A., BUELER, H., FISCHER, M., AGUZZI, A., and WEISSMANN, C. (1994):

No propagation of prions in mice devoid of PrP.

Cell 77, 967-968.

SCOTT, M., FOSTER, D., MIRENDA, C., SERBAN, D., COUFAL, F., WIILCHLI, M., TORCHIA, M.,

GROTH, D., CARLSON, G., DEARMOND, S.J., WESTAWAY, D., and PRUSINER, S.B. (1989):

Transgenic mice expressing hamster prion protein produce species-specific scrapie infectivity and amyloid

plaques.

Cell 59, 847-857.

SCOTT, M.R., WILL, R., IRONSIDE, J., NGUYEN, H.O., TREMBLAY, P., DeARMOND, S.J., and

PRUSINER, S.B. (1999):

Chapter 7- References 83

Compelling transgenetic evidence for transmission of bovine spongiform encephalopathy prions to humans.

Proc. Natl. Acad. Sci. U.S.A. 96, 15137-15142.

SHARMA, M., BURRÉ, J., and SÜDHOF, T.C. (2011b):

CSPα promotes SNARE-complex assembly by chaperoning SNAP-25 during synaptic activity.

Nat. Cell Bio. 13, 30-39.

SHARMA, M., BURRÉ, J., BRONK, P., ZHANG, Y., XU, W., and SÜDHOF, T.C. (2011a):

CSPα knockout causes neurodegeneration by impairing SNAP-25 function.

The EMBO J. 31, 829- 841.

SHMERLING, D., HEGYI, I., FISCHER, M., BLÄTTLER, T., BRANDNER, S., GÖTZ, J., RÜLICKE,

T., FLECHSIG, E., COZZIO, A., VON MERING, C., HANGARTNER, C., AGUZZI, A., and

WEISSMANN, C., (1998):

Expression of amino-terminally truncated PrP in the mouse leading to ataxia and specific cerebellar lesions.

Cell 93, 203-214.

SIGURDSON, C.J., and MILLER, M.W. (2003):

Other animal prion diseases.

Br. Med. Bull. 66, 199-212.

SIGURDSON, C.J., NILSSON, K.P., HORNEMANN, S., HEIKENWALDER, M., MANCO, G.,

SCHWARZ, P., OTT, D., RÜLICKE, T., LIBERSKI, P. P., JULIUS, C., FALSIG, J., STITZ, L.,

WÜTHRICH, K., and AGUZZI, A., (2009):

De novo generation of a transmissible spongiform encephalopathy by mouse transgenesis.

Proc. Natl. Acad. Sci. USA 106, 304-309.

SIGURDSON, C.J., WILLIAMS, E.S., and MILLER, M.W. (1999):

Oral transmission and early tropism of chronic wasting disease PrPres

in mule deer fawns (Odocoileus

hemionus).

J. Gen. Virol. 80, 2757-2764.

SISKOVA, Z., MAHAD, D.J., PUDNEY, C., CAMPBELL, G., CADOGAN, M., ASUNI, A., O'CONNOR,

V., and PERRY, V.H. (2010):

Morphological and functional abnormalities in mitochondria associated with synaptic degeneration in prion

disease.

Am. J. Pathol. 177, 1411-1421.

SISKOVA, Z., PAGE, A., O’CONNOR, V., and PERRY, V.H. (2009):

84 Chapter 7 - Reference

Degenerating synaptic boutons in prion disease: microglia activation without synaptic stripping.

Am. J. Pathol. 175, 1610-1621.

SMITH, K.J., and LASSMANN, H. (2002):

The role of nitric oxide in multiple sclerosis.

Lancet Neurol. 1, 232-241.

SOMERVILLE, R.A., OBERTHUR, R.C., HAVEKOST, U., MACDONALD, F., TAYLOR, D.M., and

DICKINSON, A.G. (2002):

Characterization of thermodynamic diversity between transmissible spongiform encephalopathy agent strains

and its theoretical implications.

J. Biol. Chem. 277, 11084-11089.

SOTO, C., and SATANI, N. (2010):

The intricate mechanisms of neurodegeneration in prion diseases.

Trends Mol. Med. 17, 1-22.

SPRAKER, T.R., BALACHANDRAN, A., ZHUANG, D., and O’ROURKE, K.I. (2004):

Variable patterns of distribution of PrPCWD

in the obex and cranial lymphoid tissues of Rocky Mountain elk

(Cervus elaphus nelsoni) with subclinical chronic wasting disease.

Vet. Rec. 155, 295-302.

SPRAKER, T.R., ZINK, R.R., and CUMMINGS, B.A. (2002):

Distribution of protease-resistant prion protein and spongiform encephalopathy in free-ranging mule deer

(Odocoileus hemionus) with chronic wasting disease.

Vet. Pathol. 39, 546-556.

TELLING, C.G., PARCHI, P., DEARMOND, S.J., CORTELLI, P., MONTAGNA, P., GABIZON, R.,

MASTRIANNI, J., LUGARESI, E., GAMBETTI, P., and PRUSINER, S.B. (1996):

Evidence for the conformation of the pathologic isoform of the prion enciphering and propagating prion

diversity.

Science 274, 2079-2082.

TELLING, G.C., SCOTT, M., HSIAO, K.K., FOSTER, D., YANG, S.L., TORCHIA, M., SIDLE, K.C.,

COLLINGE, J., DEARMOND, S.J., and PRUSINER, S.B. (1994):

Transmission of Creutzfeldt-Jakob disease from humans to transgenic mice expressing chimeric human-mouse

prion protein.

Proc. Natl. Acad. Sci. USA 91, 9936-9940.

Chapter 7- References 85

TELLING, G.C., SCOTT, M., MASTRIANNI, J., GABIZON, R., TORCHIA, M., COHEN, F.E.,

DEARMOND, S.J., and PRUSINER S.B. (1995):

Prion propagation in mice expressing human and chimeric PrP transgenes implicates the interactions of cellular

PrP with another protein.

Cell 83, 79-90.

TERRY, R.D., MASLIAH, E., SALMON, D.P., BUTTERS, N., DETERESA, R., HILL, R., HANSEN,

L.A., and KATZMAN, R. (1991):

Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive

impairment.

Ann. Neurol. 30, 572-580.

TOBABEN, S., THAKUR, P., FERNÁNDEZ-CHACÓN, R., SÜDHOF, T.C., RETTIG, J., and STAHL,

B. (2001):

A trimeric protein complex functions as a synaptic chaperone machine.

Neuron 31, 987-999.

TOIVOLA, D.M., TAO, G.Z., HABTEZION, A., LIAO, J., and OMARY, M.B. (2005):

Cellular integrity plus: organelle-related and protein-targeting functions of intermediate filaments.

Trends Cell Biol.15, 608-617.

TUZI, N.L., GALL, E., MELTON, D., and MANSON, J.C. (2002):

Expression of doppel in the CNS of mice does not modulate transmissible spongiform encephalopathy disease.

J. Gen. Virol. 83, 705-711.

UCHIYAMA, K., MIYATA, H., YANO, M., YAMAGUCHI, Y., IMAMURA, M., MURAMATSU, N.,

DAS, N.R., CHIDA, J., HARA, H., and SAKAGUCHI, S. (2014):

Mouse-hamster chimeric prion protein (PrP) devoid of N-terminal residues 23-88 restores susceptibility to 22L

prions, but not to RML prions in PrP-knockout mice.

PLoS One. 9, e109737.

VACCARI, G., DI-BARI, M.A., MORELLI, L., NONNO, R., CHIAPPINI, B., ANTONUCCI, G.,

MARCON, S., ESPOSITO, E., FAZZI, P., PALAZZINI, N., TROIANO, P., PETRELLA, A., DI-

GUARDO, G., and AGRIMI, U. (2006):

Identification of an allelic variant of the goat PrP gene associated with resistance to scrapie.

J. Gen. Virol. 87, 1395-1402.

van KEULEN, L.J., BOSSERS, A., and van ZIJDERVELD, F. (2008a):

TSE pathogenesis in cattle and sheep.

86 Chapter 7 - Reference

Vet. Res. 39, 24.

van KEULEN, L.J., VROMANS, M.E., DOLSTRA, C.H., BOSSERS, A., and van ZIJDERVELD, F.G.

(2008b):

Pathogenesis of bovine spongiform encephalopathy in sheep.

Arch. Virol. 153, 445-453.

van KEULEN, L.J.M., SCHREUDER, B.E.C., MELOEN, R.H., POELEN-VAN, D.B.M., MOOIJ-

HARKES, G., VROMANS, M.E., and LANGEVELD, J.P. (1995):

Immunohistochemical detection oand localization of prion protein in brain tissue of sheep with natural scrapie.

Vet. Pathol. 32, 299-308.

van RHEEDE, T., SMOLENAARS, N.M., MADSEN, O., and JONG, W.W. (2003):

Molecular evolution of the mammalian prion protein.

Mol. Biol. Evol. 20, 111-121.

VIDAL, E., MÁRQUEZ, M., TORTOSA, R., COSTA, C., SERAFÍN, A., and PUMAROLA, M. (2006):

Immunohistochemical approach to the pathogenesis of bovine spongiform encephalopathy in its early stages.

J. Virol. Methods. 134, 15-29.

WATTS, J.C., and PRUSINER, S.B. (2014):

Mouse models for studying the formation and propagation of prions.

J. Biol. Chem. 289, 19841-19849.

WEISSMANN C., and FLECHSIG, E. (2003):

PrP knock-out and PrP transgenic mice in prion research.

Br. Med. Bull. 66, 43-60.

WEISSMANN, C. (1991):

A “unified theory” of prion propagation.

Nature 352, 679-683.

WELLS, G.A., and WILESMITH, J.W. (1995):

The neuropathology and epidemiology of bovine spongiform encephalopathy.

Brain Pathol. 5, 91-103.

WELLS, G.A., HANCOCK, R.D., COOLEY, W.A., and RICHARDS, M.S. (1989):

Bovine spongiform encephalopathy: diagnostic significance of vacuolar changes in selected nuclei of the

medulla oblongata.

Chapter 7- References 87

Vet. Rec. 125, 521-524.

WELLS, G.A., SCOTT, A.C., JOHNSON, C.T., and GUNNING, R.F. (1987):

A novel progressive spongiform encephalopathy in cattle.

Vet. Rec. 121, 419-420.

WELLS, G.A., SPIROPOULOS, J., HAWKINS, S.A., and RYDER, S. (2005):

Pathogenesis of experimental bovine spongiform encephalopathy: preclinical infectivity in tonsil and

observations on the distribution of lingual tonsil in slaughtered cattle.

Vet. Rec. 156, 401-407.

WESTERGARD, L., CHRISTENSEN, H.M., and HARRIS, D.A. (2007):

The cellular prion protein (PrP(C)): its physiological function and role in disease.

Biochim Biophys Act. 1772, 629-644.

WILESMITH, J.W., WELLS, G.A., CRANWELL, M.P., and RYAN, J.B. (1988):

Bovine spongiform encephalopathy: epidemiological studies.

Vet. Rec. 123, 638-644.

WILL, R.G., IRONSIDE, J.W., ZEIDLER, M., COUSENS, S.N., ESTIBEIRO, K., and ALPEROVITCH,

A. (1996):

A new variant of Creutzfeldt-Jakob disease in the UK.

Lancet 347, 921-925.

WILLIAMS, E.S., and MILLER, M.W. (2003):

Transmissible spongiform encephalopathies in non-domestic animals: origin, transmission and risk factors.

Rev. Sci. Tech. 22, 145-156.

WILLIAMS, E.S., and YOUNG, S. (1980):

Chronic wasting disease of captive mule deer: a spongiform encephalopathy.

J. Wildl. Dis. 16, 89-98.

WILLIAMS, E.S., and YOUNG, S. (1993):

Neuropathology of chronic wasting disease of mule deer (Odocoileus hemionus) and elk (Cervus elaphus

nelsoni).

Vet. Pathol. 30, 36-45.

WILLIAMS, L., BROWN, P., IRONSIDE, J., GIBSON, S., WILL, R., RITCHIE, D., KREIL, T.R., and

ABEE, C. (2007):

88 Chapter 7 - Reference

Clinical, neuropathological and immunohistochemical features of sporadic and variant forms of Creutzfeldt-

Jakob disease in the squirrel monkey (Saimiri sciureus).

J. Gen. Virol. 88, 688-695.

WISNIEWSKI, H.M., SIGURDARSON, S., RUBENSTEIN, R., KASCSAK, R.J., and CARP, R.I. (1996):

Mites as vectors for scrapie.

Lancet 347, 1114.

WYATT, J.M., PEARSONG, R., SMERDOTN, N., GRUFFYDD-JONES, T. J., and WELLSG, A. H.

(1990):

Spongiform encephalopathy in a cat.

Vet. Rec. 126, 513.

YE, X., SCALLET, A.C., KASCSAK, R.J., and CARP, R.I. (1998):

Astrocytosis and amyloid deposition in scrapie-infected hamsters.

Brain Res. 809, 277-287.

YI, J.J., and EHLERS, M.D. (2007):

Emerging roles for ubiquitin and protein degradation in neuronal function.

Pharmacol. Rev. 59, 14-39.

ZHANG, J., JIN, B., LI, L., BLOCK, E.R., and PATEL, J.M. (2005):

Nitric oxide-induced persistent inhibition and nitrosylation of active site cysteine residues of mitochondrial

cytochrome-c oxidase in lung endothelial cells.

Am. J. Physiol. Cell Physiol. 288, 840-849.

ZLOTNIK, K., and RENIE, J.C. (1965):

Experimental transmission of mouse passaged scrapie to goats, sheep, rats and hamsters.

J. Comp. Pathol. 75, 147-157.

Chapter 8 - Acknowledgements 89

Chapter 8

Acknowledgements

90 Chapter 8- Acknowledgements

Chapter 8: Acknowledgements

Finally, I would like to thank all those people who made this thesis possible and an

unforgettable experience for me.

First of all, I would like to express my deepest sense of Gratitude (with a capital and bold g)

to my supervisor Professor Dr. Wolfgang Baumgärtner, who offered his continuous advice

and encouragement throughout the course of this thesis. I thank him for the systematic

guidance and great effort he put into training me in the scientific field. Like a charm!

I would also like to thanks my co-supervisors, Prof. Dr. Kirsten Haastert-Talini and Prof.

Dr. Gerd Bicker for the constructive co-supervision and brilliant comments and suggestions.

I would like to express my very sincere gratitude to Dr. Verena Haist for the support and

encouragement whenever I was in need throughout the project.

I place on record, my sincere thanks to Dr. Ingo Gerhauser and Dr. Ingo Spitzbarth for

their excellent guidance, caring, patience, for correcting manuscripts, and constructive

discussions. Without their guidance and persistent help this dissertation would not have been

possible.

I would like to thanks Prof. Martin H. Groschup and Dr. Christine Fast from Institute for

Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Südufer, Greifswald-

Insel Riems for marvellously supporting this project by providing the samples for electron

microscopy and immunohistochemistry, and providing me the opportunity to work in OIE

manual lab under their supervision. It was really a memorable part of my life.

My very sincere thanks to all members of the technical staff, without whose help this work

would definitely not have been possible, especially Kerstin Rohn for her selfless support,

encouragement, and love given to me during the course of this thesis. With her guidance and

courage, I was able to learn electron microscopy technique quite easily and she always helped

me in each and every step of electron microscopy. I would also like to thanks Brigitte

Chapter 8 - Acknowledgements 91

Behrens for continuously helping me in developing the electron microscopic images. I also

thank Bettina Buck, Petra Grünig, Danuta Waschke, Kerstin Schöne and Caroline Schütz for

their inestimable help during immunohistochemistry.

I would like to thank Muhammad Akram Khan, who as a good friend was always willing

to help and give his best suggestions. Many thanks to my office mates; Dr. Florian

Hansmann, Barbara Raddatz, Annika Lehmbecker and Adnan Fayyad for giving me

great atmosphere for working. I would also like to thanks all my colleagues including Ning,

Lin, Yimin, Yanyong for the great and unforgettable time that we spent together in the

Department of Pathology.

I am deeply thankful to Higher Education Commission (HEC), Pakistan and Deutscher

Akademischer Austauschdienst (DAAD), Germany for providing a scholarship and PMAS-

Arid Agriculture University, Rawalpindi, Pakistan for providing me leave opportunity to

complete my Ph.D.

I take this opportunity to express the profound gratitude from my deep heart to my beloved

parents (especially my late father Bashir Ahmad, that was the greatest lost I have during my

thesis work. May his soul rest in peace. Ameen), and my siblings for their love. I would like

to thank especially my brother and his wife, Mr. and Mrs. Dr. Tanveer Ahmad for their

continuous support – both spiritually and materially. I also like to thank my kids and niece

Ahmad Abdullah, Fatima-binte-Nadeem, Imsaal Tanvir and Wania Tanvir. I am sorry

that I missed your childhood.

Finally, I would like to thank my wife, Faiza Nadeem. She was always there cheering me up

and stood by me through the good times and bad.

ISBN 978-3-86345-286-5

Verlag: Deutsche Veterinärmedizinische Gesellschaft Service GmbH35392 Gießen · Friedrichstraße 17 · Tel. 0641 / 24466 · Fax: 0641 / 25375

E-Mail: [email protected] · Internet: www.dvg.de

Mu

ham

mad

Nad

eem

Han

no

ver

2015