+ All Categories
Home > Documents > Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter...

Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter...

Date post: 02-Aug-2019
Category:
Upload: nguyendiep
View: 219 times
Download: 0 times
Share this document with a friend
170
Experimental oral infection of goats with Mycobacterium avium subsp. hominissuis: Pathomorphological characterization of lesions in different courses of disease with special focus on cellular composition of granulomas Jan Schinköthe Hannover 2015
Transcript
Page 1: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

ISBN 978-3-86345-288-9

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

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

Jan

Sch

inkö

the H

an

no

ver

2015

Experimental oral infection of goats with

Mycobacterium avium subsp. hominissuis:

Pathomorphological characterization of lesions in

different courses of disease with special focus on

cellular composition of granulomas

Jan Schinköthe

Hannover 2015

Page 2: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2
Page 3: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2
Page 4: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

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 978-3-86345-288-9

Verlag: DVG Service GmbH Friedrichstraße 17

35392 Gießen 0641/24466 [email protected] www.dvg.de

Page 5: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

University of Veterinary Medicine Hannover

Experimental oral infection of goats with Mycobacterium avium subsp.

hominissuis: Pathomorphological characterization of lesions in different

courses of disease with special focus on cellular composition of granulomas

Thesis

Submitted in partial fulfilment of the requirements for the degree

-Doctor of Veterinary Medicine-

Doctor medicinae veterinariae

(Dr. med. vet.)

by

Jan Schinköthe

Sondershausen

Hannover 2015

Page 6: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Academic supervision: Apl.-Prof. Dr. E. Liebler-Tenorio

Institute of Molecular Pathogenesis

Friedrich-Loeffler-Institut, Jena

1. Referee: Apl.-Prof. Dr. E. Liebler-Tenorio

2. Referee: Prof. Dr. M. Ganter

Day of the oral examination: 18.11.2015

Financial support: This project was financially supported by the EMIDA-ERA-NET

project “Mycobactdiagnosis” (grant no. 031A083).

Page 7: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Results of this thesis were submitted or prepared for submission to journals

with a peer-review system:

J. Schinköthe, P. Möbius, H. Köhler, E. M. Liebler-Tenorio:

Experimental infection of goats with Mycobacterium avium subsp. hominissuis

- a model for comparative tuberculosis

Journal of Comparative Pathology (submitted 01.09.2015)

J. Schinköthe, H. Köhler, E. M. Liebler-Tenorio:

Characterization of tuberculous granulomas in different stages of progression

and associated tertiary lymphoid tissue in goats experimentally infected with

Mycobacterium avium subsp. hominissuis

Tuberculosis (submitted 02.11.2015)

Partial results of this thesis were presented at following congresses:

J. Schinköthe, H. Köhler, E. Liebler-Tenorio (2014)

Experimental infection of goats with Mycobacterium avium subsp.

hominissuis: Pathomorphological investigations

At: 2nd joint European Congress of the ESVP, ECVP and ESTP in Berlin, 27-

30.08.2014 (Poster)

Journal of Comparative Pathology, Volume 152, Issue 1, January 2015, Pages 83

(doi:10.1016/j.jcpa.2014.10.168)

J. Schinköthe, H. Köhler, E. Liebler-Tenorio (2015)

Experimentelle Infektion von Ziegen mit Mycobacterium avium subsp.

hominissuis (MAH): Charakterisierung der Zelltypen in den Läsionen

At: 58. Jahrestagung der Fachgruppe Pathologie der DVG, 7-8.03.2015, Fulda

(Lecture V 09)

Internet:http://tpg.schattauer.de/de/inhalt/aktuelleausgabe/issue/special/manuscript/2

4507/download.html

Page 8: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Jan Schinköthe, S. Fischer, P. Reinhold, H. Köhler, E. Liebler-Tenorio

Granulomatöse Enteritis und Lymphadenitis nach experimenteller Infektion

von Ziegen mit Mycobacterium avium subsp. hominissuis

At: Jahrestagung 2015 der Fachgruppe Krankheiten kleiner Wiederkäuer der DVG,

19-20.05.2015, Triesdorf (Lecture)

J. Schinköthe, H. Köhler, E. Liebler-Tenorio

Morphological characterization of granulomas in goats experimentally infected

with Mycobacterium avium subsp. hominissuis

At: European symposium on NTM, 24-27.06.2015, Borstel (Poster 3), 1st Poster

Award

J. Schinköthe, S. Fischer, P. Reinhold, P. Möbius, H. Köhler, E.M. Liebler-Tenorio

Different disease outcomes in goats experimentally infected with

Mycobacterium avium subsp. hominissuis (MAH) and their potential relevance

for zoonotic infection

At: National Symposium on Zoonoses Research 2015, 15-16.10.2015, Berlin

(Poster F 10), 1st Poster Award

Page 9: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2
Page 10: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

The acquisition of wealth is no longer the driving force in our lives.

We work to better ourselves and the rest of humanity.

-Captain Jean-Luc Picard-

Page 11: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Table of contents

Table of contents

1 Introduction ............................................................................................................ 15

2 Literature review .................................................................................................... 17

2.1 Mycobacterium avium subspecies hominissuis ............................................... 17

2.1.1 Taxonomy and identification ..................................................................... 17

2.1.2 Characteristics and ecology ...................................................................... 18

2.2 Host range of MAC and pathological and epidemiological features of

associated diseases .............................................................................................. 20

2.2.1 Humans ..................................................................................................... 20

2.2.2 Animals ..................................................................................................... 23

2.3 Reactions of the immune system to M. avium ................................................. 25

2.3.1 Entry at host sites ...................................................................................... 25

2.3.2 Innate and adaptive immune responses ................................................... 26

2.4 Experimental animal models in TB research ................................................... 29

2.4.1 Small animal models ................................................................................. 30

2.4.2 Large animal models ................................................................................. 32

3 Materials und methods .......................................................................................... 35

3.1 Experimental design of the oral infection of goats with MAH ........................... 35

3.1.1 Animals ..................................................................................................... 35

3.1.2 Characterisation of the MAH strain used for inoculation ........................... 36

3.1.3 Preparation of the inoculum ...................................................................... 37

3.1.4 Study design and inoculation .................................................................... 38

3.1.5 Necropsy and tissue sampling .................................................................. 41

3.1.6 Fixation of specimens with NBF ................................................................ 42

3.1.7 Cryoconservation ...................................................................................... 42

3.2 Methods used in manuscript 1 ......................................................................... 45

3.2.1 Macroscopic examination .......................................................................... 45

3.2.2 Histological examination ........................................................................... 45

3.2.2.1 Paraffin embedding ............................................................................. 45

3.2.2.2 Hemalum-eosin (HE) stain .................................................................. 46

3.2.2.3 Analysis of HE sections ...................................................................... 47

Page 12: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Table of contents

3.2.3 Immunohistochemical (IHC) detection of mycobacteria in FFPE .............. 48

3.2.4 Cultivation of MAH in tissue ...................................................................... 51

3.3 Methods used in manuscript 2 ......................................................................... 51

3.3.1 Selection and preparation of tissues ......................................................... 51

3.3.2 HE- and Azan staining of frozen sections ................................................. 52

3.3.3 Kinyoun stain ............................................................................................ 53

3.3.4 Immunohistochemical methods and antibodies used for IHC ................... 54

3.3.5 Performing of IHC ..................................................................................... 55

3.3.6 Image analysis of the size and cellular subsets in granulomas ................. 57

4 Manuscript 1 .......................................................................................................... 59

4.1 Summary ......................................................................................................... 60

4.2 Introduction ...................................................................................................... 60

4.3 Material and methods ...................................................................................... 62

4.4 Results ............................................................................................................ 67

4.5 Discussion ....................................................................................................... 80

5 Manuscript 2 .......................................................................................................... 84

5.1 Summary ......................................................................................................... 85

5.2 Introduction ...................................................................................................... 85

5.3 Material and methods ...................................................................................... 87

5.3.1 Tissue samples ......................................................................................... 87

5.3.2 Staining of frozen tissue sections .............................................................. 88

5.3.3 Immunohistochemistry (IHC) for cell surface markers and proliferation .... 89

5.3.4 Image analysis of the size and cellular subsets in granulomas ................. 90

5.4. Results ........................................................................................................... 92

5.4.1 Cellular composition of granulomas .......................................................... 92

5.4.1.1 Stage 1 ............................................................................................... 92

5.4.1.2 Stage 2 ............................................................................................... 92

5.4.1.3 Stage 3 ............................................................................................... 93

5.4.1.4 Stage 4 ............................................................................................... 94

5.4.1.5 Stage 5 ............................................................................................... 95

5.4.1.6 Stage 6 ............................................................................................... 95

Page 13: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Table of contents

5.4.2 Distribution of granuloma stages 1-6 in oGALT versus ILN of goats at 2-3

mpi versus 13 mpi ........................................................................................... 96

5.4.3 Ulcerative and granulomatous lesions in oGALT at 2-3 mpi .................. 97

5.5 Discussion ..................................................................................................... 113

5.6 Conclusion..................................................................................................... 119

6 Discussion ........................................................................................................... 121

6.1 Different courses of disease and lesions in goats after oral inoculation with

MAH .................................................................................................................... 121

6.2 Characterization of morphologic different granuloma stages ........................ 126

6.3 Outlook and possible directions of further research ...................................... 132

7 Summary ............................................................................................................. 134

8 Zusammenfassung .............................................................................................. 136

9 References .......................................................................................................... 139

10 Annex ................................................................................................................ 158

11 Acknowledgements ........................................................................................... 163

Page 14: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

List of abbreviations

List of abbreviations

3rd LT tertiary lymphoid tissue

ABC avidin-biotin-complex

AEC 3-amino-9-ethylcarbazole

AFB acid fast bacilli

AIDS acquired immunodeficiency syndrome

APC antigen-presenting cell

Aqua bidest. aqua bidestillata (latin)

Aqua dest. aqua destillata (latin)

BCG Bacillus Calmette-Guérin

BSA bovine serum albumin

BSL biosafety level

BW body weight

bwm bacterial wet mass

CD cluster of differentiation

CFP-10 culture filtrate protein of 10 kDa

cfu colony-forming unit

cm centimeter

Co-LN Lnn. Colici

COX cyclooxygenase

DAB 3,3'-diaminobenzidine

DCs dendritic cells

DNA deoxyribonucleic acid

DPBS Dulbecco's phosphate-buffered saline

dpi days post first inoculation

e.g. for example

ESAT-6 early secretory antigenic target 6

ESX-1 ESAT-6 system 1

FAE follicle-associated epithelium

FFPE Formalin fixed, paraffin embedded

Fig. figure

Page 15: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

List of abbreviations

g gram

GC germinal center

GIT gastrointestinal tract

GmbH Gesellschaft mit beschränkter Haftung

GPLs glycopeptidolipids

HE hemalum and eosin

HEV highendothelial venule

ICV-LN Lnn. ileocolici

ICVPP oGALT at colon next to ileocecal valve

IFN interferon

Ig immunglobulin

IHC immunohistochemistry

IL interleukin

ILN intestinal lymph nodes

IM intramuscular

IP immunoperoxidase

IPP ileal Peyer´s Patch

IS insertions sequence

IV intravenous

J-LN Lnn. jejunales

JPP jejunal Peyer´s patch

kg kilogram

KG Kommanditgesellschaft

KGaA Kommanditgesellschaft auf Aktien

LN lymph nodes

Ln./Lnn. lymphonodus/lymphonoduli

LP lamina propria mucosae

LPS lipopolysaccharide

m meter

M. Mycobacterium

MAA Mycobacterium avium subsp. avium

Page 16: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

List of abbreviations

MAC Mycobacterium avium complex

MAH Mycobacterium avium subsp. hominissuis

MAP Mycobacterium avium subsp. paratuberculosis

MAS Mycobacterium avium subsp. silvaticum

MB Middlebrook bouillon

MTC Mycobacterium tuberculosis complex

mg milligram

MGCs multinucleated giant cells

MHC major histocompatibility complex

min minute

MIRU mycobacterial interspersed repetitive units

ml milliliter

mm millimeter

mpi month after inoculation

NBF neutral buffered formalin

No./Nos. number(s)

NTM non-tuberculous mycobacteria

nsGPLs non serovar-specific GPLs

oGALT organized gut-associated lymphoid tissue

OHG offene Handelsgesellschaft

PBS phosphat-buffered saline

PCPP oGALT at the end of the proximal colon

PCR polymerase chain reaction

PGs prostaglandins

pH potentia hydrogenii(latin)

RA reference area

RCPP oGALT in the rectum

RD1 region of difference 1

RFLP restriction fragment length polymorphism

RT room temperature

ssGPLs serovar-specific GPLs

Page 17: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

List of abbreviations

subsp. subspecies

Tab. table

TB tuberculosis

TcR T-cell-receptor

TGF transforming growth factor

Th1/Th2 T-cell helper 1/2

TNF tumor-necrosis-factor

T7SS type 7 secretions systems

VNTR variable-number tandem repeat

µl microliter

µm micrometer

% percent

Page 18: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2
Page 19: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Introduction

15

1 Introduction

Defined experimental infections in goats with Mycobacterium (M.) avium subsp.

paratuberculosis (MAP) are used to study the pathogenesis of paratuberculosis,

search for biomarkers indicative of early infections and for evaluation of potential

vaccines (HINES II et al. 2007; KÖHLER et al. 2015; KRÜGER et al. 2015). A group

of goats inoculated with M. avium subsp. hominissuis (MAH) was included in the

infection trials, to evaluate the specificity of diagnostic antigens. In this group of

animals an unexpected onset of clinical signs with emaciation, elevated body

temperature and high-grade disturbed general condition was observed in 50 % of the

goats infected with MAH at 2-3 month after inoculation (mpi). These animals died

spontaneously or had to be euthanized according to the animal welfare law. The

remaining goats had a mild transient episode at the same time but remained healthy

until necropsy at 13 mpi.

MAH has been recognized as subspecies of M. avium since 2002 (MIJS et al. 2002)

and is widely distributed in the environment of humans and animals (BEHR u.

FALKINHAM III 2009; BIET u. BOSCHIROLI 2014). In recent years, MAH infections

have been increasingly recognized in humans and were characterized by chronic

granulomatous lesions predominantly of the respiratory tract (GRIFFITH et al. 2007;

BRODE et al. 2014). Chronic granulomatous lesions of the digestive tract are well

known in slaughter pigs usually without clinical symptoms (HIBIYA et al. 2010;

AGDESTEIN et al. 2012) and as isolated cases in various animal species with

clinically overt infection (HAIST et al. 2008; KRIZ et al. 2010).

In summary, a distinct disease entity presented in the goats infected with MAH. Thus,

this thesis addresses the pathomorphological alterations that were seen in these

goats with macroscopic, microscopic, immunohistologic, bacteriologic and genetic

approaches to answer the following questions:

Page 20: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Introduction

16

(i) What are the genetic characteristics of the MAH strain used for inoculation and

are these traits special with regard to other MAH serovars?

(ii) What lesions are caused by MAH in the goats with severe early disease at 2-3

mpi and in the goats without clinical signs at 13 mpi?

(iii) How is the amount and distribution of MAH in these lesions?

(iv) What cell types characterize these lesions?

The findings in goats infected with MAH will be compared to lesions described in

mycobacterial infections of other species to determine the usefulness of this infection

as comparative model for tuberculosis research.

Page 21: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

17

2 Literature review

This section gives an overview about the taxonomic position of MAH in the non-

tuberculous mycobacteria (NTM) and describes certain bacteriologic and ecologic

characteristics. Next, the host range and diseases associated with MAH are

explained and the immune responses of the host to M. tuberculosis and M. avium are

discussed. Finally, features of currently used small and large animal models in

tuberculosis (TB) research are described.

2.1 Mycobacterium avium subspecies hominissuis

2.1.1 Taxonomy and identification

Mycobacterium avium subsp. hominissuis (MAH) is an opportunistic environmental

pathogen and was first recognized as one of four subspecies of M. avium in 2002

(MIJS et al. 2002). The second and the third subspecies of M. avium are M. avium

subsp. avium (MAA) and M. avium subsp. silvaticum (MAS), both causes of avian

tuberculosis (TURENNE et al. 2007; RINDI u. GARZELLI 2014). The fourth

subspecies is Mycobacterium avium subsp. paratuberculosis (MAP), the causative

agent of Johne´s disease, a chronic granulomatous enteritis in ruminants (CLARKE

1997; RINDI u. GARZELLI 2014; KRÜGER et al. 2015). Genetic analysis revealed

that MAP, MAA and MAS evolved from an ancestral MAH by deletion and acquisition

of genomic sequences (RINDI u. GARZELLI 2014). M. avium, 8 other species and

species that are not further determined, establish the so called Mycobacterium avium

complex (MAC, RINDI u. GARZELLI 2014). The MAC itself is classified into the large

group of NTM currently comprising more than 150 species, which can be broadly

determined by gene sequencing of conserved targets like 16S rRNA (TORTOLI

2012; I. M. ORME u. ORDWAY 2014). However, the resolution of this method is not

sufficient and can be enhanced when sequencing other genetic targets, e.g. hsp65,

which distinguishes pathogenic subspecies of M. avium or the internal transcribed

spacer element (BEHR u. FALKINHAM III 2009). Apart from sequence based typing,

testing for the presence or absence of certain insertion sequences (IS) of M. avium

subspecies is very helpful and allows direct detection of the organism (BEHR u.

Page 22: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

18

FALKINHAM III 2009). Usually, MAH contains a polymorphic multibanded IS1245

pattern, multiple copies of IS1311 without IS901 elements and grows at a wide

temperature range of 24 to 45 °C (MIJS et al. 2002; TURENNE et al. 2007; RINDI u.

GARZELLI 2014).

2.1.2 Characteristics and ecology

The mycobacterial cell wall (Fig. 2.1) is a complex structure mainly composed of

peptidoglycans, arabinogalactans and mycolic acids followed by a capsule of various

proteins and lipids. These cell wall components confer the acid fastness, which is

being utilized for visualization of mycobacteria with acid fast stains, e.g Ziehl-Neelsen

or Kinyoun (BRENNAN 2003; KIESER u. RUBIN 2014). In M. tuberculosis, type 7

secretions systems (T7SS) are responsible for the transport of proteins through the

mycobacterial cell wall. These proteins are associated with granuloma formation and

the escape of mycobacteria into the cytosol of macrophages (ABDALLAH et al. 2007;

STOOP et al. 2012). Five different T7SSs exist in M. tuberculosis. The early

secretory antigenic target 6 (ESAT-6) system 1 (ESX-1) is best studied since it

secretes ESAT-6 and culture filtrate protein of 10 kDa (CFP-10) both associated with

virulence and granuloma formation. The region of difference 1 (RD1) which encodes

ESX-1 is absent in Mycobacterium bovis Bacille Calmette-Guerin (BCG). It confers

the lower virulence of BCG (ABDALLAH et al. 2007). RD1 is partially or completely

deleted in M. avium species (STOOP et al. 2012). However, the lack of these

virulence factors seems to be compensated by special cell wall components of M.

avium, the glycopeptidolipids (GPLs). GPLs are non-covalently attached to the outer

cell wall layer of NTM, but not of M. tuberculosis and M. leprae (HONDA et al. 2015).

They can be divided into non serovar-specific GPLs (nsGPLs), that are present in

many NTM species and serovar-specific GPLs (ssGPLs) that are solely produced by

M. avium and allow the typing of currently 31 distinct serotypes of M. avium (HONDA

et al. 2015).

Page 23: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

19

Fig. 2.1 Mycobacterial cell wall: 1, outer lipids; 2, mycolic acid; 3, polysaccharides

(arabinogalactan); 4, peptidoglycan; 5, plasma membrane; 6, liparabinomannan

(LAM); 7, phosphatidylinositol mannoside; 8, glycopeptidolipids (GPLs); 9, cell wall

skeleton

The original work has been modified.

"Mycobacterial cell wall diagram" by Y tambe - Y tambe's file.

Licensed under CC BY-SA 3.0, http://creativecommons.org/licenses/by-sa/3.0/

https://commons.wikimedia.org/wiki/File:Mycobacterial_cell_wall_diagram.png#/media/File:Mycobacter

ial_cell_wall_diagram.png

Serotypes 4-6, 8-11 and 21 are classified as MAH (MÖBIUS et al. 2006; HIBIYA et

al. 2011). ssGPLs in particular are highly immunogenic components necessary for

intracellular survival, the regulation of the production of pro- and anti-inflammatory

cytokines, biofilm formation and are therefore part of the species-specific

pathogenesis (SWEET u. SCHOREY 2006; SCHOREY u. SWEET 2008;

Page 24: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

20

MUKHERJEE u. CHATTERJI 2012; HONDA et al. 2015). In addition, GPLs in

general can resist lysosomal degradation, accumulate in macrophages and can

trigger adjacent macrophages when shed via exosomes (BHATNAGAR et al. 2007).

The ability of M. avium to form biofilms is connected with the survival in water supply

systems due to adherence in pipes. Thus persistence in drinking water systems can

occur and is considered as major risk factor for dissemination in human households

in the United States (FALKINHAM 2013). This lead to the recommendation that tap

water should not be used for sinus irrigation when patients suffer from chronic

rhinosinusitis, because it may harbor M. avium (TICHENOR et al. 2012; FALKINHAM

2013). After intake into the environment M. avium can resist diverse harsh

environments mainly due to its impermeable, lipid-rich cell wall. It was frequently

isolated from soil, particular peat-rich soil which promotes mycobacterial growth due

to the low acidic pH of 3-5 (FALKINHAM 2009, 2013). Recent data from Germany

reported detection of MAH primarily in dust and soil, while biofilms and water were

free (LAHIRI et al. 2014). Taken together MAH and other NTM species are widely

distributed in the environment, however with regional differences.

2.2 Host range of MAC and pathological and epidemiological features of

associated diseases

Discrepancies in the reviewed literature have been found concerning the designation

of MAC, M. avium or MAH as the etiologic agent of diseases mainly through the

following reasons: (1) MAH exists as distinct subspecies only since 2002 (MIJS et al.

2002) and reports of diseases caused by NTM before this time were virtually

described as MAC or M. avium and (2) even nowadays, where sophisticated

discrimination methods are available, hospital labs do not routinely distinguish

between M. avium subspecies (BEHR u. FALKINHAM III 2009).

2.2.1 Humans

MAC infections gained great importance during the emergence of the acquired

immunodeficiency syndrome (AIDS) epidemic in the 1980´s. AIDS patients with

disseminated granulomatous disease in a variety of organs were increasingly

Page 25: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

21

recognized, among them predominantly homosexual man with a history of

intravenous and non-intravenous drug abuse and concomitant infections (DAMSKER

u. BOTTONE 1985; ROTH et al. 1985; WOLINSKY 1992). Particularly the

gastrointestinal tract (GIT), intestinal lymph nodes (ILN), liver, spleen and almost all

other organs can be affected with fever, emaciation and diarrhea as common

symptoms. Histopathologic examination revealed aggregates of macrophages filled

with numerous AFB frequently replacing parts of preexisted tissue at these sites

(ROTH et al. 1985; ARIS et al. 2011).

MAC was considered as the most common NTM species (WOLINSKY 1992) and this

feature continued to date particular in developed countries (HOEFSLOOT et al. 2013;

ORME u. ORDWAY 2014). Nowadays, MAH is considered as most relevant, since

nearly 70% of AIDS patients die after untreated MAH infection (IGNATOV et al.

2012).

Also immunocompetent people, especially children, older people (>60 years) and a

group of middle-aged, postmenopausal women with a slender body phenotype are

affected by MAC-associated diseases (GRIFFITH et al. 2007; KARTALIJA et al.

2013). Among the MAC species, predominantly MAH causes granulomatous neck

lymphadenitis in children (BRUIJNESTEIJN VAN COPPENRAET et al. 2008) and

granulomatous lesions of the respiratory tract in the other patient groups (TRAN u.

HAN 2014). Children develop primarily neck lymphadenitis most likely through oral

uptake of MAH when playing with soil (DESPIERRES et al. 2012).

Three patterns of respiratory diseases can be differentiated: (i) hypersensitivity

pneumonitis, also known as hot-tub lung, that is associated with aerosols which

derive from household water or shower; (ii) tuberculosis-like pulmonary disease

which is frequently connected to chronic obstructive pulmonary disease, cigarette

smoking or prior TB; and (iii) nodular bronchiectasis (Fig. 2.2) frequently seen in older

females with the aforementioned slender body phenotype (WEISS u. GLASSROTH

2012; ORME u. ORDWAY 2014).

Page 26: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

22

Fig. 2.2 Nodular bronchiectasis in a CT scan also known as “Lady Windermere

syndrome”.

"Lady windermere syndrome ct" by Samir at en.wikipedia. Licensed under CC BY 3.0

http://creativecommons.org/licenses/by-sa/3.0/

https://commons.wikimedia.org/wiki/File:Lady_windermere_syndrome_ct.JPG#/media/File:Lady_winde

rmere_syndrome_ct.JPG

Little is known about the histopathological picture of pulmonary MAH infection. In an

autopsy-based study of 11 deceased patients almost all had an organized

granulomatous inflammation predominantly necrotizing and to a lesser extent non-

necrotizing or were diffusely consolidated with scattered AFB (O’CONNELL et al.

2012). Those were bronchocentrically concentrated, accompanied by bronchiectasis

with 73% of cases having cavities and 36% nodules. Microbiologic data revealed

MAC without further differentiation in eight and M. abscessus in three of the patients

(O’CONNELL et al. 2012).

In general, current data suggest an overall increase of NTM infections in developed

countries, with MAC species as leading cause (HOEFSLOOT et al. 2013; BRODE et

Page 27: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

23

al. 2014; ORME u. ORDWAY 2014). However, reliable data about incidence rates

are rare, since NTM infections are usually not mandatory. One exception is

Queensland, Australia, that documented a rise in incidence rates of NTM infections

from 4.85/100.000 in 1999 to 5.7/100.000 in 2005. Interestingly, this rise was

concomitant with an almost constant incidence rate of MTC infections: 2.5/100.000

(1999) and 2.6/100.000 (2005) (BRODE et al. 2014). Brode and colleagues showed,

that this phenomenon is present in almost all developed countries and discussed

different aspects of cross immunity, socio-economic factors and improved diagnostic

techniques as responsible for this trend.

2.2.2 Animals

Predominantly pigs are affected and a variety of other species, e.g. horses, cats,

dogs, cattle, wild ruminants and zoo animals, as indicated from recently published

reports (GLAWISCHNIG et al. 2006; MÖBIUS et al. 2006; HAIST et al. 2008; KRIZ et

al. 2010; AGDESTEIN et al. 2012; MORAVKOVA et al. 2013; AGDESTEIN et al.

2014; KLANG et al. 2014). Local granulomatous infections of the GIT, ILN (Fig. 2.3)

and liver accompanied by clinically inapparent disease predominated in young pigs

(< 6 month). Lesions were usually detected at slaughter, while systemic infections

were rare. Exsudative lesions with multinucleated giant cells (MGCs), macrophages,

lymphocytes, neutrophils and eosinophils predominated. Necrosis and calcification

were seen in lesions of ILN, while fibrotic organization was less obvious

(AGDESTEIN et al. 2012). In the other species, affected individuals are usually older

(> 1 year) and have clinically overt disease with emaciation, weight loss and a

prolonged phase of diarrhea that often leads to euthanasia due to animal welfare

reasons.

Page 28: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

24

Fig. 2.3 Multifocal yellow-white foci (arrows) with a dry and crumbly consistency in a

mandibular LN of a pig infected with M. avium.

The photograph is provided with the kind permission of Dr. André Vallant, Landratsamt Dingolfing-

Landau; Abteilung Veterinärwesen.

Shedding of numerous MAH via feces was also mentioned during phases of

diarrhea. At necropsy, animals had frequently disseminated disease with (pyo)-

granulomatous inflammation in GIT, ILN and a variety of organs. The lesions were

characterized by extensive areas of caseous necrosis with calcification surrounded

by epitheloid macrophages and MGCs with numerous AFB (GLAWISCHNIG et al.

2006; HAIST et al. 2008; KRIZ et al. 2010).

Epidemiological data describing the current status of MAH infections in animals are

scarce since they are not notifiable or reportable. A more satisfactory situation exists

for MAP, which is notifiable in Germany (BMEL 2015) and is listed in the Terrestrial

Animal Health Code of the Organization for Animal Health (SEVILLA et al. 2015).

Page 29: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

25

Without considering MAP infections, where significant epidemiological data exist,

MAH infections are mostly reported in case studies and merely few research articles

are available. A recent study of hunted wild boars in Spain revealed that 16,8 % of

animals were infected with NTM species and among these 11% belonged to the

species M. avium (GARCÍA-JIMÉNEZ et al. 2015). The lack of epidemiological data

is explained by a bias in tissue sampling. Most studies are currently focused on

sampling respiratory tissue to decipher epidemiological aspects of TB, while M.

avium infections are primarily seen in the GIT (BIET u. BOSCHIROLI 2014).

2.3 Reactions of the immune system to M. avium

Most studies which describe host immune response to M. avium are predominantly

derived from experimental infections in murine models. Frequently serotype 1

(BERMUDEZ et al. 1992; KIM et al. 1998; SANGARI et al. 2001; PETROFSKY u.

BERMUDEZ 2005; HAUG et al. 2013) was used for inoculation of animals which

nowadays is considered to be MAA (DHAMA et al. 2011; HIBIYA et al. 2011) and

little is known about MAH, even though both subspecies are closely related (BIET u.

BOSCHIROLI 2014). The following paragraphs are primarily focused on the

pathogenesis of MAA, MAH and M. intracellulare. When appropriate, comparisons to

the pathogenesis of tuberculosis and paratuberculosis are drawn.

2.3.1 Entry at host sites

Uptake of M. avium can occur via the respiratory or the alimentary tract in humans

and animals. Soft tissue and skin infections in humans are also reported, but are

usually iatrogenic or caused by trauma (HONDA et al. 2015). In the respiratory tract,

M. avium infects bronchial epithelial cells via binding to fibronectin on integrin

receptors of epithelial cells (HONDA et al. 2015). Fibronectin-associated binding to

epithelial cells, predominantly M cells, was also reported in MAP infections (SECOTT

et al. 2004). It is generally accepted that the preferred site of MAP entry are M cells

overlying organized gut-associated lymphoid tissue (oGALT, MOMOTANI et al. 1988;

SIGURDARDÓTTIR et al. 2001). However, experimental studies in mice with M.

Page 30: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

26

avium serovar 1 suggested that MAA primarily invades the intestine via enterocytes

(SANGARI et al. 2001).

After overcoming this first epithelial barrier, macrophages and dendritic cells (DCs)

are the primary target cells for M. avium. Toll-like receptors, complement receptors,

mannose receptors, cluster of differentiation (CD) 14, Fc receptors or fibronectin

receptors are exploited to gain access into these cell types via ssGPLs (ROCCO u.

IRANI 2011; HONDA et al. 2015). Once inside macrophages or DCs, MAH resides in

phagosomes where they actively inhibit the phagolysosome maturation, similar to M.

tuberculosis (CARDONA u. IVANYI 2011; IGNATOV et al. 2012) or if not, even

survive the detrimental environment inside the phagolysosome (ROCCO u. IRANI

2011).

2.3.2 Innate and adaptive immune responses

To M. tuberculosis

If phagocytic cells fail to kill mycobacteria, the pro-inflammatory cytokines interleukin

(IL) 12, IL-1β and tumor necrosis factor (TNF) and various chemokines are released

(FLYNN et al. 2011; O'GARRA et al. 2013). TNF-α and chemokines attract

neutrophils, monocyte-derived macrophages, NK cells and γδ T lymphocytes to the

site of infection (PETERS u. ERNST 2003). The continuous immigration of

macrophages results in the development of an early granuloma and is thought to

promote M. tuberculosis replication (EHLERS u. SCHAIBLE 2012). DCs that contain

mycobacteria are responsible for the induction of adaptive immunity. They migrate to

draining lymph nodes (LN) and prime naïve CD4+ and CD8+ T lymphocytes, which

reside in paracortical regions of LN, by secretion of IL-12 (PETERS u. ERNST 2003;

FLYNN et al. 2011). Studies revealed that this is a delayed process, since priming of

T lymphocytes started usually 12-21 days post infection (WOLF et al. 2008;

O'GARRA et al. 2013). As explanation for this delay was proposed, that DCs infected

with M. tuberculosis are impaired in the ability to migrate from the lung to the draining

LN, while this is not the case for DCs that have phagocytosed bacilli in apoptotic

bodies (BLOMGRAN u. ERNST 2011).

Page 31: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

27

After priming by DCs, antigen-specific, activated T lymphocytes migrate back to the

site of granulomatous inflammation. Th1 polarized CD4+ T lymphocytes secrete IFN-γ

which activates macrophages, while CD8+ T lymphocytes are involved in direct

cytotoxic killing of infected macrophages. At this stage, the granuloma becomes

organized with infected epitheloid macrophages at the center surrounded by a layer

of lymphocytes (EHLERS u. SCHAIBLE 2012). Concomitantly, regulatory T

lymphocytes and anti-inflammatory cytokines like IL-10 and TGF-β released by

macrophages limit tissue pathology by downregulation of pro-inflammatory

responses and establish a balanced response between bacterial killing and host

survival (FLYNN et al. 2011; O'GARRA et al. 2013).

The role of B lymphocytes and humoral immunity were neglected for a long time

even though the presence of B lymphocytes within granulomas is well known

(PETERS u. ERNST 2003). However, in recent years it became clear that they have

a major influence in granuloma development and clinical outcome of human TB

(ULRICHS et al. 2004; ULRICHS u. KAUFMANN 2006). Ulrichs and colleagues

suggested that not the granuloma center itself, but rather the lymphocytic layer in the

periphery is the primary battlefield between host and mycobacteria. This assumption

was based on their findings that granulomas had T lymphocyte-rich and B

lymphocyte-rich areas resembling tertiary lymphoid tissue (3rd LT). Mycobacteria in

CD68+ cells were frequently associated with B lymphocyte-rich areas which indicate

antigen presentation in this environment (ULRICHS et al. 2004; ULRICHS et al.

2005). The B lymphocyte-rich compartments had many proliferative cells and were

therefore interpreted as germinal centers (GC). Patients that had well developed 3rd

LT in the periphery of granulomas were in the asymptomatic latent phase of TB,

while patients with active disease had cavitary lesions without 3rd LT (ULRICHS et al.

2005). Thus, compartimentalization of lymphocytes into 3rd LT confers protective

immunity and highlights the importance of B lymphocytes and humoral immunity in

the interaction between mycobacteria and the host (ULRICHS u. KAUFMANN 2006;

CHAN et al. 2014).

The CD8+ T lymphocytes were not found in the granulomatous rim surrounding the

necrotic core, but rather in the T lymphocyte-rich areas of the lymphocytic rim. This

Page 32: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

28

makes their relevance for necrosis formation by killing of infected macrophages

questionable (ULRICHS u. KAUFMANN 2006). To answer the question what factors

drive necrosis or apoptosis in tuberculous lesions, eicosanoids are increasingly

investigated for their role in cell death of infected macrophages (BEHAR et al. 2010).

When tissue is damaged, phospholipids from the cell wall are converted by

phospholipases for the production of arachidonic acid (TIZARD 2012). Arachidonic

acid is further processed by lipoxygenases to produce leukotrienes and anti-

inflammatory lipoxins (e.g. LXA4) or cyclooxygenases (COX) which produce pro-

inflammatory prostaglandins (PGs), e.g. PGE2. These arachidonic acid derivatives

are termed eicosanoids. They influence cell death, amount of TNF-α and indirectly

TB outcome (BEHAR et al. 2010; KAUFMANN u. DORHOI 2013).

Apoptosis is thought to be detrimental for mycobacteria, since mycobacteria in

apoptotic macrophages are cross presented to DCs resulting in an effective immune

response restricting mycobacterial growth and limiting tissue pathology (BEHAR et al.

2010; DIVANGAHI et al. 2010). Therefore, mycobacteria can actively block apoptosis

of macrophages (VELMURUGAN et al. 2007) and influence eicosanoid pathways.

Attenuated M. tuberculosis induces PGE2 which prevents necrosis by repairing

mitochondrial membranes damaged by mycobacteria (DIVANGAHI et al. 2010).

Generally speaking, mycobacteria can shift the eicosanoid pathways in their favor

towards apoptosis. Virulent M. tuberculosis induces increased levels of LXA4 by

inhibiting COX-2, which lead to decreased levels of PGE2 finally resulting in necrosis

of infected macrophages (BEHAR et al. 2010). Thus, necrosis is beneficial for

mycobacteria, since it results in uncontrolled release of mycobacteria into the

extracellular milieu, promotes massive proliferation of mycobacteria and

dissemination while immune cells are absent (BEHAR et al. 2010; RAMAKRISHNAN

2012; MATTY et al. 2015).

To M. avium

After entry into macrophages, M. avium infections progress similar as described for

M. tuberculosis and may result in formation of organized (non-) necrotizing

granulomas or diffuse granulomatous disease (O’CONNELL et al. 2012). There are,

Page 33: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

29

however, differences in the involvement of certain cell types (HAUG et al. 2013;

ORME u. ORDWAY 2014). IFN-γ producing CD4+ T lymphocytes, but not CD8+ or γδ

T lymphocytes are required for an effective immunity against M. avium in mice. This

is supported by the findings in AIDS patients that succumb to disseminated M. avium

infections when CD4+ T lymphocytes are decreasing (PETROFSKY u. BERMUDEZ

2005; HAUG et al. 2013). M. avium infections vary greatly in their outcome

depending on the virulence of strains used for infection (HAUG et al. 2013). While

granulomas in mice infected with M. tuberculosis show no evidence of necrosis

(LENAERTS et al. 2015), aerosol infection of mice with virulent M. avium can result in

uncontrolled bacterial growth and severe necrosis of granulomas (BENINI et al.

1999). As in TB, CD4+ Th1 lymphocytes and an intact IL-12/IFN-γ axis are required

for the development of necrosis, while conflicting results exist about the role of TNF-

α. Some investigations observed the development of necrotic granulomas in TNF-

receptor deficient mice (FLORIDO et al. 2002), while others reported the opposite

(ORME u. ORDWAY 2014). On the other hand, excessive apoptosis induced by

biofilm formation was reported. Analysis of macrophages after exposure to MAH

biofilms resulted in rapid apoptosis and hyper stimulation of macrophages in vitro

(ROSE u. BERMUDEZ 2014). In summary, it is less clear what drives apoptosis or

necrosis when compared to data from TB studies.

The role of B lymphocytes in M. avium infections is almost unknown and studies that

report 3rd LT in association with granulomas does not exist. However, B lymphocytes

in addition to CD4+ and CD8+ T lymphocytes are required for protective immunity

against M. avium in mice. The authors (FATTORINI et al. 1999) suggest that not

antibody production, but rather antigen presentation is the role of B lymphocytes in

this experimental model.

2.4 Experimental animal models in TB research

The granuloma is the hallmark of TB composed of different cell types like epitheloid

macrophages, MGCs, lymphocytes and neutrophils. With further granuloma

development necrosis, mineralization, fibrosis and cavity formation are variably

present. This demonstrates that there is not one type of granuloma, but rather a wide

Page 34: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

30

heterogeneity of morphologically and functionally different granulomas which can be

found even in the same host (EHLERS u. SCHAIBLE 2012; LENAERTS et al. 2015).

Therefore, animal models should aim to reproduce the heterogeneity of lesions and

disease progression as it occurs in human TB (BASARABA 2008). However, there is

not a single animal model that fully recapitulates the pathology of human tuberculosis

(EHLERS u. SCHAIBLE 2012). In the following, specific features of currently used

animal models are described.

2.4.1 Small animal models

The majority of experimental studies in TB research are done in mice. This is due to

the fact that a variety of genetically modified strains, several knock-out technics and

a plethora of immunological and immunohistochemical tools exist for studying

interactions between host and pathogen. Thus, a lot of fundamental insights have

been gained. However, most mice do not develop human TB-like lesions with

necrotic and mineralized granulomas or cavity formation (VILAPLANA u. CARDONA

2014; LENAERTS et al. 2015). Merely the C3HeB/FeJ mouse strain develops

necrotic granulomas (DRIVER et al. 2012).

In addition, there is the issue of housing conditions of the mice. Usually, mice are

kept in research facilities at temperatures of 19-22 °C which are suboptimal for their

physiology (KARP 2012). As small mammals they are adapted to arid environments,

temperatures of 30-32 °C are rather preferred. When the establishment of the

thermoneutral zone is impaired by the housing regimen, chronic cold stress occurs

that alters a wide variety of immunological effects in mice (KOKOLUS et al. 2013).

Thus, conclusions drawn from mouse studies have to be considered with great care,

particular in cancer and infectious disease research (KARP 2012; KOKOLUS et al.

2013; ENG et al. 2015).

The guinea pig model overcomes some of the constraints that occur in mice models

of TB. In particular, they are used for drug testing, since they develop necrotic and

calcified granulomas allowing the study of drug distribution and response of M.

tuberculosis in different microenvironments (LENAERTS et al. 2007; BASARABA

Page 35: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

31

2008). However, the feature of cavitary lesions is also lacking in this species

(LENAERTS et al. 2015).

The zebrafish model provides live imaging of cellular processes in the early phase of

granuloma development due to the use of transparent zebrafish larvae infected with

M. marinum and revolutionized the understanding of granuloma formation. Early

granulomas with epitheloid macrophages, necrotic and hypoxic regions developed in

the absence of any adaptive immunity and displayed early spread of mycobacteria by

inducing secondary granulomas (DAVIS u. RAMAKRISHNAN 2009). Also the above

mentioned influences of eicosanoids on inflammatory pathways, especially TNF-

mediated pathways, were convincingly demonstrated in this model (TOBIN et al.

2012). With regard to these findings, the former common view of the granuloma as

host protective entity shifted towards replication and exploitation of the granuloma by

virulent mycobacteria (RAMAKRISHNAN 2012; MATTY et al. 2015).

In addition and of great importance, all above mentioned species do not undergo a

phase of latency, but rather develop a progressive disease and finally die

(BASARABA 2008; VAN RHIJN et al. 2008; GUIRADO u. SCHLESINGER 2013).

Latent TB is defined as "evidence of immunological sensitization by mycobacterial

proteins, in the absence of clinical signs and symptoms of active disease” (BARRY et

al. 2009). Compared to active disease, granulomas in latent TB show minor

inflammation with prominent fibrosis and mineralization (LENAERTS et al. 2015). An

animal model capable to establish granuloma heterogeneity during a phase of

latency would be highly desirable, since almost 2 billion humans are latently infected

with TB and fail to completely eliminate the infection, and thus are the major source

for developing active disease when immunity wanes (KAUFMANN u. DORHOI 2013).

During active disease, individuals have sterile granulomas, caseous necrotic

granulomas with variable bacillary load and liquefactive cavitary lesions with

numerous replicating mycobacteria (BARRY et al. 2009).

In non-human primates, particular in the macaque model, development of the full

spectrum of TB pathology with latency and active disease, granuloma heterogeneity

and even cavitary lesions can be found (CAPUANO et al. 2003; LENAERTS et al.

2015). However, ethical issues, high costs and the restricted availability of animals

Page 36: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

32

have to be considered when applying this animal model (VAN RHIJN et al. 2008;

LENAERTS et al. 2015).

2.4.2 Large animal models

In particular, ruminants gained access into TB research for different advantageous

reasons. Cattle and goats are natural outbred hosts for M. bovis and M. caprae, that

are both closely related to M. tuberculosis (CRAWSHAW et al. 2008; SHARPE et al.

2010; PESCIAROLI et al. 2014; WATERS et al. 2014). Additionally, both species are

of zoonotic potential (BIET et al. 2005; RODRÍGUEZ et al. 2009) and few studies

reported about M. tuberculosis infections of cattle and goats (AMENI et al. 2011;

KASSA et al. 2012). The primary mode of infection is the respiratory route via

inhalation of mycobacteria and like in humans minimal doses are sufficient (VAN

RHIJN et al. 2008). Furthermore, epidemiological aspects of TB transmission can be

recapitulated, since livestock animals are usually kept in herds which allow the study

of infectious disease dynamics within a defined population (LANZAS et al. 2010).

Special traits of human anatomy are absent in small animal species and are only

present in large animal species like cattle, goat and swine (VILAPLANA u.

CARDONA 2014). One example that illustrates the significance of this aspect is the

presence of intralobular septae in human lungs and large animal species. This

feature primarily influences the tendency of granuloma encapsulation in experimental

settings and mimics human pulmonary TB where most granulomas are restricted to

certain regions of the lung sections (GIL et al. 2010). The question, if the

encapsulation process is relevant or not for the outcome of TB, can be studied only in

large animal models (CARDONA 2015).

The major advantage of ruminants as animal models is the establishment of a wide

range of morphologically different lesions that are found in natural and experimentally

induced TB and closely mimic human pathology (WANGOO et al. 2005; VAN RHIJN

et al. 2008; DOMINGO et al. 2009; DOMINGO et al. 2014; LENAERTS et al. 2015).

Different macroscopic lesions can be semi-quantitatively assessed at necropsy by

careful examination of affected organs using scoring systems that allow comparison

between different studies (VORDERMEIER et al. 2002; DOMINGO et al. 2014). This

Page 37: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

33

can be improved by microscopic staging of different granulomas. Wangoo and

colleagues established a scheme for granuloma staging in bovine LN: stage 1-

clusters of epitheloid macrophages, stage 2 - variable, thin encapsulated granulomas

with minimal necrosis, stage 3 - completely encapsulated with central caseous and

mineralized necrosis and stage 4 - marked fibrosis, with extensive multicentric

caseous and mineralized necrosis (WANGOO et al. 2005). The authors concluded

that staging of granulomas might be helpful to recognize differences between certain

Mycobacteria species or changes in immunity. Meanwhile, many research groups

utilized this approach which allows comparisons between multiple studies, different

methods and the conclusions drawn (LIEBANA et al. 2007; LIEBANA et al. 2008;

SANCHEZ et al. 2011; ARANDAY-CORTES et al. 2013; PALMER et al. 2015). Due

to these advanced analytic tools, it is not surprising that both species are widely used

as animal models in TB vaccine research aiming to control the infection in the

respective species and also for safety and efficacy studies of human vaccines

(VORDERMEIER et al. 2006; DE VAL PEREZ et al. 2011; WATERS et al. 2012;

DOMINGO et al. 2014).

Particularly, TB infection in goats raised attention in recent years, since they are of

small size in comparison to cattle which enables smaller housing capacities and

allows performance of state of the art imaging techniques (DE VAL PEREZ et al.

2011; GONZALEZ-JUARRERO et al. 2013). Especially the possibility to perform

magnetic resonance imaging and computed tomography is of notice which enhances

the assessment of lesion distribution in the lung and allows the study of granuloma

development. Unfortunately, despite of all efforts, there is up to date no TB vaccine

that fully prevents infections and lesion establishment (DOMINGO et al. 2014).

Finally, it is worth mentioning that cattle and goats are one of few species that

develop cavitary lesions during active disease (DOMINGO et al. 2014; LENAERTS et

al. 2015). Prior to onset of active disease, a variable, subclinical phase occurs even

with the possibility of clearance of infection in some animals (VAN RHIJN et al.

2008). Thus, both species are able to mimic the clinically important phase of latency

which was recognized in recent years and exploited by some studies even though no

Page 38: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Literature review

34

inducible model of latent bovine or caprine tuberculosis currently exists (VAN RHIJN

et al. 2008; DOMINGO et al. 2009).

Page 39: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

35

3 Materials und methods

This chapter is structured as follows: paragraph 3.1 describes the animal experiment

in detail, while paragraphs 3.2 and 3.3 describe the methods used in the two

manuscripts submitted. Paragraph 3.2 deals with the histopathological

characterization of lesions and detection of mycobacteria and paragraph 3.3

describes the investigations of selected lesions to further characterize their cellular

composition.

3.1 Experimental design of the oral infection of goats with MAH

3.1.1 Animals

Thirty-one goats („Thüringer Waldziegen“) derived from a herd without a history of

mycobacteriosis where used in this study. Twenty-six animals were of male gender, 4

of female gender and 1 hermaphrodite. All goats were taken at the age of 8-19 days

to the animal facility of the FLI, Jena. At this time fecal samples were collected and

cultured, but no MAH or MAP was detected. Additionally, monitoring for other

infectious agents (Salmonella, Mycoplasma and Pasteurella) was performed and

resulted negative. The ten control animals and the 21 goats designated for

inoculation with MAH were kept in separate animal rooms. The rooms had deep

straw bedding. Ad libitum feed of hay and a permanent access to water were given at

any time. Initially the goat kids received commercial milk replacement (Denkamilk

Capritop, Denkavit Futtermittel GmbH, Warendorf, Germany). Until 10 weeks of life

(12kg body weight) this was stepwise changed to concentrated feed (Alleinfuttermittel

für Ziegenmastlämmer, Landhandelsgesellschaft eG, Schmölln, Germany) and hay

until the end of the trial. The male goats were castrated between 7 and 8 weeks of

life and received metamizole (40mg/kg, intramuscularly (IM), WDT, Germany) for 3

days. A prophylactic therapy against coccidia (Baycox®, 20mg/kg, Bayer,

Leverkusen, Germany) was performed at the 3rd and 4th month of life. This was

accompanied by supplementary administration of vitamin B1 (Vitamin B1-Hevert®,

200 mg/animal, IM, Hevert Nussbaum, Germany) at the 3rd, 5th, 8th and 11th month of

life together with the treatment against endo- and ectoparasites with doramectin

Page 40: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

36

(Dectomax®; 3 mg/kg, IM, Pfizer, Berlin, Germany) at the 5th, 8th and 11th month of

life.

In concordance with the national Animal Welfare Act, this animal experiment was

approved by the animal ethical committee of Thuringia and the Thuringian Regional

Office for Food Safety and Consumer Protection (Permit Number: 04-002/12). All

experiments were done in containment of biosafety level (BSL) 2 under supervision

of the authorized institutional agent for animal protection. During the entire study,

every effort was made to minimize suffering.

3.1.2 Characterisation of the MAH strain used for inoculation

The investigations on the genetic traits of MAH are described in this paragraph and

were kindly performed by Dr. Petra Möbius.

Bacterial strain

Strain 09MA1289 was isolated from a lymph node of a slaughtered pig in Baden

Württemberg in Germany with suspicion of mycobacteriosis in 2009 on Stonebrink

medium with Pyruvat, PACT and Loewenstein-Jensen medium with Glycerol and

PACT (Bioservice Waldenburg, Germany) without Mycobactin J. The isolate was

maintained and propagated in Middlebrook 7H9 broth containing glycerine and 10%

OADC (MB, Becton Dickinson, Heidelberg, Germany).

DNA extraction

Genomic DNA was prepared by the cetyltrimethylammonium bromide method as

described (VAN SOOLINGEN et al. 1991).

Identification

Species and subspecies determination was done by Mycobactin independency

during cultivation, examination for the presence of IS1245 (GUERRERO et al. 1995),

the absence of IS900 (ENGLUND et al. 1999), the absence of IS901 (KUNZE et al.

Page 41: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

37

1992), and the presence of flanking region of IS901 FR300 without the IS901

element (NISHIMORI et al. 1995).

Genotyping

Restriction fragment length polymorphism (RFLP) analysis based on the insertion

sequence IS1245 was conducted as recently published (MÖBIUS et al. 2006), but

using the short probe of IS1245 (JOHANSEN et al. 2005) to prevent cross

hybridization between IS1245 and IS1311. Strain 09MA1289 was further

characterized by mycobacterial interspersed repetitive units-variable-number tandem

repeat (MIRU-VNTR) typing and using foreseeable polymerase chain reaction (PCR)

product sizes for data analysis (THIBAULT et al. 2007; RADOMSKI et al. 2010). In

detail, different numbers of tandem repeat sequences were detected by PCR

targeting specific loci of the M. avium genome (MIRU-VNTR Loci 292, X3, 25, 47, 3,

7, 10, 32). Results were arranged in the same order and profiles were called INMVs

(THIBAULT et al. 2007).

Detection of potential virulence markers

The presence of ISMpa1 was examined by PCR using the primers P2 and P3

(OLSEN et al. 2004). The presence of nsGPL genes (gtfA, rtfA, mtfC) and ser2

genes (mdhtA, merA, mtfF) involved in the synthesis of GPLs in the cell wall of M.

avium was investigated using published primers (JOHANSEN et al. 2009).

3.1.3 Preparation of the inoculum

The inoculum was prepared essentially as described elsewhere with modifications

(KÖHLER et al. 2015). Bacterial inoculum stocks were prepared from batch cultures

in MB. After centrifugation of the stocks, bacterial wet mass was adjusted to 10 mg

per dose using PBS. The bacterial inoculum of MAH strain 09MA1289 amounted to

0.78 - 4.1 x 109 colony forming units (cfu) per dose.

Page 42: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

38

3.1.4 Study design and inoculation

All goats were evenly distributed based on body weight and origin of the sire. Thus

seven goats each to be infected with MAH were assorted into three groups housed in

the same animal room. Inoculation was performed by Dr. H. Köhler. Twenty-one

goats received at the age of 10-21 days, 10 times, every 2-4 days the inoculum with

the milk replacer. The total inoculation doses yielded 2.13 x 1010 cfu MAH per goat.

The 10 control goats were sham inoculated with milk replacer only. Necropsy dates

were scheduled for 4, 7 and 13 mpi. Necropsy of 10 control animals was scheduled

at the end of the trial together with infected animals at 13 mpi.

Unexpectedly, 12 of 21 MAH infected goats died spontaneously or had to be

euthanized according to the animal welfare law (Tab. 3.1). Goat No.1 had to be

euthanized 28 days after inoculation (dpi) and revealed a severe pleuropneumonia

with multiple abscesses at necropsy. Goat No. 2 died spontaneously due to a

volvolus 30 dpi. Goats 3-11 died spontaneously or had to be euthanized for animal

welfare reasons between 2-3 mpi after a phase of severely protracted disease. Goat

No.12 was necropsied 4 mpi as originally scheduled. The remaining 9 goats survived

and were regularly necropsied with the 10 control goats at 13 mpi (Tab. 3.1, Fig. 3.1).

Page 43: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

39

Fig. 3.1 Timeline of necropsies of MAH-inoculated and control goats. The grey

arrows indicate animals that died separately from the other MAH infected goats. The

red arrows indicate the groups of MAH infected goats, while the green arrow

indicates the group of control goats.

Page 44: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

40

Tab. 3.1 Overview of goats inoculated with MAH and controls

Goat No.

Goat No. in

manus-cript

Gen-der

Age at start of

the study (in d)

Weight at start of the study (in kg)

Age at start/end

of in-oculation

(in d)

Age at ne-

cropsy (in d)

Weight at ne-cropsy (in kg)

Ne-cropsy (in dpi)

M1 M2

1 - - ♂ 11 5,0 13/39 41 6,4 28

2* - - ♂ 16 6,6 18/44 48 11,6 30

3 1 - ♂ 8 4,6 10/36 76 10,8 66

4* 2 - ♀ 13 7,0 15/41 86 10,8 71

5 3 1 ♂ 12 4,2 14/40 90 12,4 76

6 4 - ♂ 13 7,4 15/40 93 15,6 78

7 5 2 ♀ 13 3,2 13/39 91 10,4 78

8 6 3 ♂ 16 6,0 18/44 96 14,9 78

9 7 4 ♂ 11 4,4 13/39 96 13,9 83

10 9 - ♂ 10 6,2 12/38 96 16,4 84

11 8 5 ♂ 10 5,0 12/38 97 15,8 85

12 - - H 14 5,4 16/42 133 27,0 117

13 10 7 ♂ 19 7,6 21/47 386 38,8 365

14 11 8 ♂ 15 5,8 17/43 382 36,4 365

15 12 9 ♂ 13 6,0 15/41 387 37,6 372

16 13 10 ♂ 11 4,4 13/39 385 35,6 372

17 14 11 ♂ 11 6,6 13/39 396 39,6 383

18 15 12 ♂ 14 6,8 16/42 399 44,9 383

19 16 13 ♂ 12 4,6 14/40 399 37,3 385

20 17 14 ♂ 13 5,4 15/41 405 41,7 390

21 18 15 ♀ 12 3,6 14/40 404 32,8 390

22 19 - ♂ 17 8,0 - 381 45,1 362

23 20 - ♂ 15 6,8 - 381 51,3 362

24 21 - ♀ 12 4,0 - 383 33,9 369

25 22 - ♂ 11 5,2 - 404 38,2 391

26 23 - ♂ 11 5,8 - 404 45,2 391

27 24 - ♂ 10 4,6 - 404 42,0 392

28 - - ♂ 14 5,4 - 385 45,7 369

29 - - ♂ 10 6,4 - 404 39,4 392

30 - - ♂ 13 6,0 - 377 41,0 362

31 - - ♂ 12 5,6 - 383 35,7 369

*died spontaneously; dpi, days post first inoculation; M1, manuscript 1; M2,

manuscript 2; H, hermaphrodite

Su

rviv

ors

C

on

trol a

nim

als

D

ece

ase

d a

nim

als

Page 45: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

41

3.1.5 Necropsy and tissue sampling

Necropsies started with the sedation of goats with xylazine 2% (Rompun®, 0.25

mg/kg IM, Bayer, Leverkusen, Germany). This was subsequently followed by

administration of ketamine-hydrochlroride (Ketamin 10%®, 2.5 mg/kg, intravenously

(IV), Intervet, Unterschleißheim, Germany) and diazepam (Faustan®, 0.5 mg/kg, IV,

AWD, Radebeul, Germany) to induce deep anesthesia and was maintained by

ketamine-hydrochloride adjusted to depth of anesthesia. The reason behind this

procedure was to ensure preservation of intestinal tissues, particular the mucosa, on

predefined intestinal segments to generate high quality sections later on. To achieve

this aim, the abdominal cavity was opened and predefined small intestinal segments

were selected and loops ligated. Loops at each localization were either filled with 4 %

neutral buffered formalin1 (NBF) or with cold phosphate buffered saline2 (PBS) by

injection into the ligated small bowel lumen. Four jejunal localizations, either ligated

two meters apart (Tab. 3.2, goats No. 1-12) or three meters apart (Tab. 3.2, goats

No. 13-31), two jejunal Peyer´s patches (JPP) from the proximal (JPP-p) and distal

(JPP-D) jejunum, and two segments of the ileal Peyer´s patch (IPP) 80 cm (IPP-1)

and 30 cm (IPP-2) from the entrance of the ileocecal valve were prepared. Without

any delay the goat was euthanized with pentobarbital-sodium (Release®, 20 ml/goat,

WDT, Garbsen, Germany). After this, the intestines were cut at the duodenum and

rectum, detached from the mesentery and placed on a table. The complete intestinal

length was measured. Prior ligated intestinal segments were measured with regard to

their localization and were cut out of the intestine. A variety of other intestinal

segments and organs were sampled: duodenum, cecum, oGALT in the colon next to

ileocecal valve (ICVPP), at the end of the proximal colon (PCPP) and in the rectum

(RCPP), ansa centralis, and descending colon (when macroscopic alterations were

present). The remaining bowel was opened at the mesenteric attached site and color,

consistency and amount of feces were recorded.

1 own production, see annex

2 own production, see annex

Page 46: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

42

Then the ingesta were rinsed off and the mucosa was investigated for macroscopic

lesions. Additional JPP with lesions were collected, termed, JPP-variable (JPP-v),

and their position in the intestine were measured. The ILN were examined and

samples were taken from proximal and distal jejunal lymph nodes (J-LN), ileocolic

lymph nodes (ICV-LN) and colonic lymph nodes (Co-LN). The necropsy ended with a

complete sampling of a variety of representative organs (Tab. 3.2). Figure 3.2 gives

an overview of the sampling of intestinal segments and ILN.

Dr. H. Köhler harvested tissues with sterile scissors and forceps at comparable sites

for cultural isolations of mycobacteria (Tab. 3.2).

3.1.6 Fixation of specimens with NBF

All NBF filled and ligated intestinal segments were opened at the mesenteric

attachment and pinned flat on a styrofoam with the mucosal surface upwards. Then

samples were submerged in NBF. The other organs and lymph nodes were directly

placed into NBF filled containers. All specimens were fixated at least for 24 h in a

ratio of 1:10 tissue to NBF.

3.1.7 Cryoconservation

For cryoconservation, PBS filled intestinal segments were opened at the mesenteric

attachment, rinsed, placed on a thin slice of liver tissue and snap frozen for 20-60

seconds at -70 °C in 2-methylbutane (Carl Roth GmbH & Co. KG, Karlsruhe,

Germany). The remaining tissues were directly snap frozen at -70 °C. All tissues

were wrapped in aluminium foil, labelled and stored at -80 °C.

Page 47: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

43

Fig. 3.2 Overview of intestinal sites where samples were collected for histopathology

(red arrows) and for cyroconservation (green arrows). Duo, duodenum; Jej, jejunum;

Cae, cecum; JPP, oGALT in jejunum; IPP, oGALT in the ileum; ICVPP, oGALT in the

colon next to ileocecal valve (ICVPP), at the end of the proximal colon (PCPP) and in

the rectum (RCPP), modified from Krüger (2014).

Page 48: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

44

Tab. 3.2 Overview of tissue samples fixated in NBF, snap frozen and collected for

cultural isolation of mycobacteria.

Localisation NBF-fixation Cryoconservation

Cultural isolation of mycobacteria

1 duodenum x x x

2 jejunum 1 x x x

3 jejunum 2 x x

4 jejunum 3 x x x

5 jejunum 4 x x x

6 JPP proximal x x x

7 JPP distal x x

8 JPP-v x x

9 IPP 1 x x

10 IPP 2 x x

11 Caecum x x

12 ICVPP x x x

13 PCPP x x x

14 ansa centralis x x

15 colon altered x x

16 rectum x x

17 Lnn. jej. cranialis x x x

18 Lnn.jej. caudalis x x 2x

19 Lnn. ileocolici x x x

20 Lnn. colici x x

21 Lnn. hepatici x x x

22 liver x x x

23 tonsills x x x

24 Ln. ing.superf. x x Lnn. cerv.superf.

25 Lnn. retrophar. x x x

26 thymus x

27 spleen x x x

28 pancreas x

29 kidney x x

30 adrenals x

31 lung x

32 Ln. mediastinalis x

33 heart x

34 aorta x

35 rumen x

36 abomasum x

37 bone marrow x

Page 49: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

45

3.2 Methods used in manuscript 1: Pathomorphological examinations and

detection of MAH in tissues

3.2.1 Macroscopic examination

A thorough visual examination of all specimens was performed and all alterations

were recorded with special emphasis on the gut and the oGALT and ILN.

3.2.2 Histological examination

3.2.2.1 Paraffin embedding

NBF fixed tissues were trimmed and placed into embedding cassettes (Engelbrecht,

Edermünde, Germany). Prior to trimming extensively mineralized ILN were

decalcified with HCL-EDTA decalcifying solution (Richard Allen Scientific, Microm

International, Germany) for 12 h. Tissues were rinsed under cold tap water for at

least one hour to remove the fixative. The dehydration and paraffin (Paraffin type 1,

Microm International GmbH, Thermo Fisher Scientific, Walldorf, Germany) infiltration

was performed with the tissue processor Tissue-Tek® VIP® 6 (Sakura Finetek, Inc.,

Torrance, USA). The protocol is summarized in table 3.3.

Tab. 3.3 Protocol for paraffin embedding in the tissue processor Tissue-Tek® VIP® 6

Solvents Temperature (in °C) Duration (in h)

Ethanol 70% ( 2-times) 40 1

Ethanol 96% ( 3-times) 40 1

Isopropanol ( 2-times) 40 1

Xylene (2-times) 40 1

Paraffin 1+2 63 Each 1

Paraffin 3+4 63 Each 1,5

The processed tissues were placed into metal dishes, embedded in paraffin (Paraffin

type 6, Thermo Fisher Scientific GmbH, Walldorf, Germany) in a paraffin dispensing

center Histocentre 2 Shandon (Thermo Scientific GmbH, Walldorf, Germany).

Formalin fixed, paraffin embedded (FFPE) consecutive sections of 1.5 to 2.0 µm

thickness were generated on a rotary microtome (HM355S, Thermo Fisher Scientific,

Page 50: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

46

Walldorf, Germany) and collected on chrome alum gelatin3 coated slides (StarFrost,

Engelbrecht, Edermünde, Germany) or electrostatically loaded slides (Süssefrost

Plus, Süsse Labortechnik, Gudensberg, Germany). The manufactured sections were

dried in a warming cabinet (Mikrobiologischer Brutschrank B6200, Kendro Laboratory

Products GmbH, Langenselbold, Germany) for 24-48 h at 37 °C.

3.2.2.2 Hemalum-eosin (HE) stain

Sections were deparaffinized starting with a warming up period at 60 °C for 30 min in

a warming cabinet. The remaining paraffin was removed by rinsing twice in xylene for

two minutes. Then sections were rehydrated in denatured 96 % alcohol and in

denatured 70% alcohol twice for two minutes and five minutes in distilled water.

Sections were stained for 25 minutes in Mayer´s hemalum4, rinsed in distilled water

and blued under cold tap water for 10 min. The counterstain was performed with 1%

eosin5 for four minutes. After a renewed rinse in tap water, the sections were

dehydrated and differentiated (Tab. 3.4) This process ended with cover slipping in

Canada balsam (Carl Roth GmbH & Co. KG, Karlsruhe, Germany). The HE stain

stains nuclei blue and the cytoplasm in red.

3 own production, see annex

4 own production, see annex

5 own production, see annex

Page 51: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

47

Tab.3.4 Dehydration and differentiation of HE stain

Solvents Duration (in min)

denatured ethanol 70% ( 2-times) short

denatured ethanol80% ( 2-times) short

denatured ethanol 96% ( 2-times) 2

carbolic xylene6 3

xylene 2

3.2.2.3 Analysis of HE sections

All specimens that were collected during necropsy were examined for the presence

of histologic lesions in HE stained sections. All lesions were analyzed for distribution,

severity and localization of inflammatory cells. Lesions were summarized as mild (+):

small granulomatous or lymphocytic infiltrates without influencing the tissue

morphology, moderate (++): inflammation influenced the tissue morphology or severe

(+++): partial or complete disruption of tissue morphology due to a variable infiltration

of inflammatory cells. The overall assessment of each localization depended on the

most severe lesions detectable in the section. The distribution of lesions was

assessed as: a) focal - <3 lesions per section, multifocal - >3 lesions per section and

diffuse - lesions without obvious separation. A special scheme for granuloma staging

was adopted from a previously published study (WANGOO et al. 2005) and is

summarized in Tab. 3.5.

6 own production, see annex

Page 52: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

48

Tab. 3.5 Characterization of granulomas in oGALT and lymph nodes

Granuloma stage

Cellular composition Necrosis and mineralization

Fibrosis

1 (Initial)

epitheloid macrophage cluster, ± MGCs, interspersed and encircling lymphocytes,

single neutrophils

no necrosis -

2 (Solid) epitheloid macrophages, ± MGCs, interspersed with

lymphocytes and neutrophils

minimal necrosis and/or

mineralization

incomplete,

thin: -/+

3 (Monocentric necrosis and

mineralization)

± epitheloid macrophages, ± MGCs, lymphocytes and

neutrophils, surround necrosis and/or mineralization

monocentric caseous necrosis

and/or mineralization

variable,

- / +++

4 (Multicentric necrosis and

mineralization)

± epitheloid macrophages, ± MGCs, lymphocytes and

neutrophils, surround necrosis and/or mineralization

multicentric caseous necrosis and/or mineralization

variable,

- / +++

5 (Fibrotic organization)

minimal cellular infiltrate

fibrosis > necrosis and mineralization

+++

MGCs, multinucleated giant cells; -, not present; ±, missing or present at variable

number.

3.2.3 Immunohistochemical (IHC) detection of mycobacteria in FFPE

IHC is used to identify bacterial or host antigens. Antigen-antibody binding is

visualized by light microscopy. To visualize MAH in FFPE tissue, sections were

placed on electrostatically loaded slides and the indirect immunoperoxidase (IP)

method was performed. Prior to this, FFPE sections were deparaffinized beginning

with a warming phase in a warming cabinet at 60 °C for 30 min. The remaining

paraffin was removed by rinsing in xylene followed by rehydration (Tab. 3.6). The

endogenous peroxidase was blocked by incubation of sections in 0.3 % H2O2 (Merck

Schuchardt OHG, Hohenbrunn, Germany) in methanol for 25 minutes at room

temperature (RT). This was followed by triple rinsing in Tris-PBS-buffer7 for 5 minutes

7 own production, see annex

Page 53: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

49

Tab.3.6 Removing of paraffin and rehydration of consecutive sections for IHC

Solvents Duration (in min)

xylene( 2-times) 3

ispropanol ( 2-times) 3

denatured ethanol 96% ( 2-times) 3

denatured ethanol 70% 2

denatured ethanol 50% 5

distilled water 5

each. For antigen retrieval tissue was digested in trypsin. For this, trypsin (Sigma-

Aldrich Chemie GmbH, Steinheim, Germany) and CaCl2 (Merck KGaA, Darmstadt,

Germany) were mixed and adjusted to pH 7.8. Sections were incubated for 20

minutes at 37 °C in a waterbath (Memmert GmbH & Co. KG, Schwabach,

Germany).The reaction was stopped by rinsing in cold tap water for 10 minutes. This

was followed by rinsing for five minutes with Tris-PBS-buffer and subsequent

placement of sections in Shandon coverplates (Thermo Fisher Scientific GmbH,

Nidderau, Germany). To avoid unspecific antibody binding, sections were incubated

with 10% of normal goat serum diluted in 12,5 % bovine serum albumin (BSA,

SERVA Electrophoresis GmbH, Heidelberg, Germany) in Dulbecco’s PBS8 (DPBS)

for 20 min at RT. Then a polyclonal rabbit anti-MAP serum (Dako, Glostrup,

Denmark) diluted 1:4000 in DPBS (pH 7.4) containing 12.5% BSA was used as

primary antibody and sections were incubated for 60 minutes at RT. After triple

rinsing in Tris-PBS buffer the incubation with horseradish peroxidase-conjugated

goat anti–rabbit IgG as secondary antibody (Dianova, Hamburg, Germany) was done

for 60 minutes at RT. Again a triple rinsing in Tris-PBS buffer followed with removal of

sections from coverplates.

For visualization of the antigen-antibody reaction, 3-amino-9-ethylcarabazol-

formamid (AEC) was used as chromogen. Sections were incubated for 20 minutes in

8 own production, see annex

Page 54: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

50

AEC solution9 at RT. Since AEC generates an intense red precipitate this feature was

used to differentiate between unspecific brown pigments in the cytoplasm of

macrophages in intestinal and LN sections. The final steps were a short rinse in

distilled water, a counterstaining for 90 seconds in Mayer´s hemalum, bluing under

cold tap water for 10 min and cover slipping with Kaiser´s glycerin gelatin (Merck

KGaA, Darmstadt, Germany).

To guarantee specificity of each antigen-antibody reaction positive and negative

controls were used. A MAH suspension was injected into a muscle tissue to generate

a tissue block for positive control slides. The above mentioned polyclonal anti-MAP

was cross reactive against MAH in the positive control slides. This positive control

was replaced during the infection trial by a FFPE tissue block of an IPP with many

MAH in the granulomatous lesions of goat No.11. The cultivation followed by PCR of

adjacent IPP verified the presence of MAH in tissue. A consecutive section of the

positive control was incubated with a primary antibody against unrelated antigen

(Brachyspira hyodysenteria) as negative control.

Analysis of IHC detection of MAH

All intestinal, oGALT, ILN, tonsils, liver, liver LN, superficial inguinal LN and lung

specimens, as well as tissues with granulomatous inflammation were investigated by

IHC for the presence of MAH by light microscopy (Laborlux S, Leitz GmbH, Wetzlar,

Germany) according to a adopted scheme (KRÜGER et al. 2015). The adopted

scheme additionally considered the presence of mycobacterial material in caseous

necrosis and mineralization that was seen in granulomas (Tab. 3.7). To clarify the

specificity of labelling in necrotic material, for each section with caseous necrosis a

consecutive section was incubated with a primary antibody against unrelated bacteria

(Brachyspira hyodysenteria). Only labeling with the polyclonal anti-MAP serum

resulted in specific granular labeling in necrosis and mineralization.

9 own production, see annex

Page 55: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

51

Tab. 3.7 Scoring of labeled MAH and mycobacterial material in tissue sections by

IHC

Quantity Distribution

None < 2 MAH per section, no labelling in n and m

Single (a) <20 % EM/ MGC with single MAH and/or single labeling of mycobacterial material in n+m

Few (b) 20-50 % EM/MGC with 1-10 MAH and/or predominantly single to moderate labeling of mycobacterial material in n+m

Many (c)

50-75 % EM/MGC with >10 MAH and < 50% with uncountable MAH and/or predominantly moderate to extensive labeling of mycobacterial material in n+m

Numerous (d) >75 % EM/MGC with >10 MAH and > 50% with uncountable MAH and/or predominantly extensive labeling of mycobacterial material in n+m

n, necrosis; m, mineralization; EM, epitheloid macrophages; MGC, multinucleated

giant cells

3.2.4 Cultivation of MAH in tissue

Cultivation and the molecular-biological identification of MAH from tissues that were

sampled during necropsy, was done in the workgroup “Mycobacteria” headed by Dr.

Köhler. All investigated tissues were collected from sites adjacent to the specimens

used for histopathological examination. Detailed analysis of growth kinetics for each

specimen were performed, however, in the manuscript merely the statement

“presence or absence of mycobacteria in tissue” was included.

3.3 Methods used in manuscript 2: IHC labeling of cell surface markers,

proliferative cells, endothelial cells and staining of collagen and acid fast

bacteria in granulomas

3.3.1 Selection and preparation of tissues

The cellular composition of granulomas and granulomatous lesions that were

predominantly found in oGALT and ILN was the objective of manuscript 2 (Tab. 3.8).

Page 56: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

52

Tab. 3.8 Origin of tissues used (goats, localizations)

Time of necropsy 2-3 mpi 13 mpi

Tissue

localizations

JPP 6 13

IPP 5 11

ILN 5 12

Total No. of goats sampled 5 9

Identification No. of goats 5, 7-9, 11 13-21

Snap frozen tissues of JPP, IPP and ILN had been collected and stored as described

in section 3.1.7. Frozen samples were cut at -29 to -24 °C with cooled foreceps and a

razor blade in a cryostate (Figocut 2800E, Leica Instruments GmbH, Wetzlar,

Germany) to tissue blocks of approximately 1.5 cm x 0.5 cm x 0.5 cm for oGALT and

2.0 cm x 1.0 cm x 0.5 cm for ILN. Tissue blocks were attached with freezing medium

(NEG 50TM, Thermo Fisher Scientific, Walldorf, Germany) to the object holder.

Consecutive sections of 5 µm in thickness were generated and collected on chrome

alum gelatin coated slides. Sections were dried at RT overnight and subsequently

stored at - 20 °C until further use.

3.3.2 HE- and Azan staining of frozen sections

Sections were dried at RT for 60 min, fixed in NBF for 5 minutes and then rinsed in

distilled water for 5 minutes.

HE-stain

To analyze granulomatous lesions and perform granuloma staging all specimens

were stained with HE. A new granuloma stage was added to table 3.5 and resulted in

an extended granuloma scheme (Tab. 3.9). Five minutes staining with Mayer´s

hemalum was followed by further steps as described in section 3.2.2.2.

Page 57: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

53

Azan stain

The Azan stain was used for demonstration of collagen fibers. After rinsing in distilled

water, sections were stained for 13 minutes in azocarmine B solution10 at 60 °C.

Renewed rinsing in distilled water was followed by differentiation in aniline alcohol11

for 3 minutes, in acetic alcohol12 for 1 minute and in phosphomolybdic acid (Merck

KGaA, Darmstadt, Germany) for 15 minutes. Sections were rinsed in distilled water

and further stained for 13 minutes in an aniline blue/orange G solution13. Sections

were rinsed in distilled water, followed by incubation in absolute ethanol for 3 minutes

twice and for 2 minutes in xylene twice. The procedure ended with coverslipping of

sections in Canada balsam.

3.3.3 Kinyoun stain

This method was used without heating of sections for visualization of AFB and was

performed with a commercially available staining kit following the manufacturer´s

instructions (BD-TB-Stain Kit K, Omni Lab, Markkleeberg, Germany). Sections were

air dried for 60 minutes and then differentiated for 4 minutes in TB Carbolfuchsin KF,

rinsed with distilled water, decolored with decolorizer for 30 seconds, rinsed with

distilled water and finally counterstained for 1 minute in TB Brilliant Green K. The

staining was finished by incubation for 5 minutes in xylene and coverslipping of

sections in Canada balsam.

Numbers of AFB were semi- quantitatively assessed at x 1000 magnification in a

reference area (RA) of 2359 µm2 on representative sections (Tab. 3.11).

10

own production, see annex

11 own production, see annex

12 own production, see annex

13 own production, see annex

Page 58: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

54

Tab. 3.9 Characterization of granulomas in oGALT and lymph nodes

Granuloma stage

Cellular composition Necrosis and mineralization

Fibrosis

1 (Initial)

epitheloid macrophage cluster, ± MGCs, interspersed and

encircling lymphocytes, single neutrophils

no necrosis -

2 (Solid) epitheloid macrophages, ± MGCs, interspersed with

lymphocytes and neutrophils

minimal necrosis and/or

mineralization -

3 (Monocentric necrosis and

mineralization)

± epitheloid macrophages, ± MGCs, lymphocytes and

neutrophils surround necrosis and/or mineralization

monocentric caseous necrosis

and/or mineralization

variable,

- / +++

4 (Multicentric necrosis and

mineralization)

± epitheloid macrophages, ± MGCs, lymphocytes and

neutrophils surround necrosis and/or mineralization

multicentric caseous necrosis

and/or mineralization

variable,

- / +++

5 (Fibrotic organization)

minimal cellular infiltrate

fibrosis > necrosis and mineralization

+++

6 (Extensive caseous necrosis)

many lymphocytes, interspersed epitheloid macrophages and

neutrophils,

irregular necrosis with minor

mineralization -

MGCs, multinucleated giant cells; -, not present; ±, missing or present at variable

number

3.3.4 Immunohistochemical methods and antibodies used for IHC

Both the indirect IP method (described in section 3.2.3) and the avidin-biotin complex

(ABC) method were used. For the latter, a biotinylated secondary antibody with

enzyme labeled ABC was used. Avidin is a tetravalent glycoprotein with a strong

affinity for biotin. This connection is exploited by binding of biotin to the secondary

antibody and the labeling enzyme (horseradish peroxidase). After incubation of the

biotinylated secondary antibody with the primary antibody a complex of avidin and

biotinylated enzyme is added. The enzyme catalyzes a chromogen and an amplified

signal occurs in form of an insoluble precipitate.

Page 59: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

55

To examine the cellular composition of different lesions and granulomas, monoclonal

antibodies against CD4, CD8, CD25, TcR1-N24 (δ chain), CD79α, CD68 and MHC-II

were used (Tab. 3.10). The monoclonal antibody Ki-67 and the polyclonal rabbit anti-

human van Willebrand Factor (vWF) were used for detection of proliferative cells,

respectively for detection of endothelial cells, capillaries and larger vessels. Table

3.10 summarizes cell types, epitopes, clones, dilutions and information about the

suppliers of the primary and secondary antibodies used. The ABC method was used

for the primary antibodies against CD25 and CD79α and the indirect IP method for

the primary antibodies against CD8, TcR1-N24 (δ chain), CD68, MHC-II, Ki-67 and

vWF. CD4 was enhanced by using a commercially available Boost Kit SignalStain®

Boost IHC Detection Reagent (New England Biolabs GmbH, Frankfurt, Germany)

following the manufacturer´s protocol.

3.3.5 Performing of IHC

Frozen sections were air dried for 60 minutes at RT, fixated in aceton (CD4, CD8,

TcR1-N24(δ chain), CD79α, MHC-II and vWF), in -20 °C methanol/aceton (1:2) for

CD25 or in NBF for Ki-67 for 10 min. The sections were than air dried for 10 minutes,

surrounded by a hydrophobic circle using a felt-tip pen (Dako Pen, Dako, Hamburg,

Germany) and rehydrated twice in DPBS for 5 minutes each. The NBF fixed sections

were immediately rinsed in DPBS. To avoid unspecific binding to the tissue, sections

were incubated with a blocking serum (inactivated, 1:10 diluted with BSA/DPBS) for

20 minutes at RT in humid chambers (Carl Roth GmbH & Co. KG, Karlsruhe,

Germany). The blocking serum was from the same species as the secondary

antibody was. After removal of the blocking serum, sections were incubated with the

primary antibody for 60 minutes at RT in humid chambers. After rinsing off the

primary antibody, sections were transferred into DPBS for 5 minutes, 3-times. This

was followed by blocking of the tissue intrinsic peroxidase by incubation in 0.06 %

phenylhydrazine solution14 at 37 °C in a water bath for 40 minutes. After renewed

rinsing in DPBS, sections were incubated with the secondary antibodies for 60 min at

14

own production, see annex

Page 60: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

56

RT in humid chambers.

Tab. 3.10 Antisera used for IHC

target/ cells

epitope/ antigen

clone dilution supplier secondary antibody

method

T helper cells

CD4 GC1A 1:50

VMRD, Pullman, USA, distrib. Labor Diagnostik Leipzig,

Germany

Boost Kit15

indirect immuno-

peroxidase (IP)

cytotoxic T cells

CD8 CC63 un-

diluted

European Cell Culture Collection,

Salisbury, UK

sheep anti-mouse IgG

16

indirect IP

γδ T cells TcR1-N24 (δ chain)

GB21A 1:400 VMRD sheep anti-

mouse IgG16

indirect IP

activated T cells

CD25 (IL-2Rα)

CACT 116A

1:500 VMRD biotin. goat anti-mouse

IgG17

, ABC18

avidin-biotin

complex

B cells, plasma

cells CD79α HM57 1:200

AbD Serotec, Düsseldorf, Germany

biotin. goat anti-mouse

IgG17

, ABC18

avidin-biotin

complex

macro-phages

CD68 EBM11 1:50 Dako, Glostrup,

Denmark sheep anti-

mouse IgG16

indirect IP

MHC-II MHC-II H42A 1:4000 European Cell

Culture Collection, Salisbury, UK

sheep anti-mouse IgG

16

indirect IP

prolifer-ating cells

Ki-67 MIB-1 1:200 Dako, Glostrup,

Denmark sheep anti-

mouse IgG16

indirect IP

endo-thelial cells

Van Willebrand

Factor ---

1:800, 1:2000*

Dako, Glostrup, Denmark

goat anti-rabbit IgG

19

indirect IP

* dilution of 1:800 used in oGALT and 1:2000 used in ILN

15 SignalStain® Boost, New England Biolabs GmbH, Frankfurt, Germany

16 NA931V, GE Healthcare Europe GmbH, Freiburg

17 GE Healthcare Europe GmbH, Freiburg, Germany

18 Vector Laboratories, Inc., Burlingame, USA; distributed by Biologo, Konshagen, Germany

19Jackson Immunoresearch; distributed by Dianova, Hamburg, Germany;

Page 61: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

57

After rinsing in DPBS, the ABC Kit Vectastain® (Vector Laboratories, Inc.,

Burlingame, USA; Biologo, Konshagen, Germany) was used for the clones

CACT116A and HM57. For each section 1 ml of DPBS was mixed with each 20 µl of

reagent A, respectively reagent B and was left to stand for 30 minutes at RT.

Sections were incubated for 30 minutes with ABC and finally rinsed in DPBS. To

visualize the peroxidase conjugated secondary antibodies and peroxidase

conjugated avidin-biotin complex, 3,3’-diaminobenzidine (DAB) was used as

chromogen. After short rinsing in distilled water, 1-2 droplets of 0.01 % osmic acid

solution20 were placed for 20 seconds resulting in blackening of the precipitate.

Counterstaining was done for 10 to 15 minutes with 2 % methylene green solution21,

with the exception for Ki-67 and vWF reactions that were counterstained in Mayer´s

hemalum for 20 seconds. After rinsing in distilled water sections were cover slipped

with Kaiser`s glycerin gelatin. To verify specificity of the reactions, positive controls

that included all cell types to be visualized were carried along in each reaction. As

negative control, a consecutive section served by replacing the primary antibody with

an antibody of the same isotype against unrelated antigen (Brachyspira

hyodysenteriae).

3.3.6 Image analysis of the size and cellular subsets in granulomas

Measurement of granuloma areas in sections was used as estimate of granuloma

size. This was performed with Image J for all granulomas of all stages in selected

JPP, IPP and ILN. Granulomas were measured by manual tracing of their outline in

HE-stained sections. Stages 1 and 2 were measured at x 100 magnification, stage 3

on x 1 or x 50 magnification depending on size, stages 4 and 5 x 1 magnification and

stage 6 on x 1 or x 50 magnification depending on size. Magnification of factor 1 was

achieved by scanning whole slides with a flatbed scanner (Epson V700, Seiko Epson

Corporation, Suwa, Japan).

20own production, see annex

21own production, see annex

Page 62: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Materials and methods

58

Numbers of CD4+, CD8+, CD25+, TcR1-N24+, CD79α+, CD68+, Ki-67+ cells,

endothelial cells and neutrophils were semi-quantitatively assessed at x 400

magnification in a RA of 2359 µm2 measured by an ocular grid. Cell numbers were

converted to scores (Tab. 3.11). MGCs were differentiated as small MGCs (<10

nuclei) and large MGCs (>10 nuclei). Representative images were captured using a

Leica DMR microscope (Leica, Germany) equipped with an Olympus DP71 video

camera (Olympus, Germany) and imaging software (Cell F, Version 2.6, Olympus,

Germany).

Tab. 3.11 Semi-quantitative assessment of different immunohistochemically labeled

cell types, neutrophils and AFB

score - (+) + ++ +++ ++++

none scattered single few many numerous

number of lymphocytes, plasma cells, proliferating

cells/RA

0 1-2 3-8 9-14 15-20 >20

number of macrophages/RA 0 1-2 3-6 7-10 11-15 >15

Number of AFB 0 1-3 4-15 16-50 50-200 >200

RA, reference area (2359 µm2)

Page 63: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

59

4 Manuscript 1

Experimental infection of goats with Mycobacterium avium subsp. hominissuis

- a model for comparative tuberculosis research

Authors: Jan Schinköthe, Petra Möbius, Heike Köhler, Elisabeth M. Liebler-Tenorio

Affiliations:

Jan Schinköthe, DVM

Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health,

Naumburger Str. 96a, 07743 Jena, Germany

e-mail: [email protected]

P. Möbius, Dr. rer. nat.

Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health,

Naumburger Str. 96a, 07743 Jena, Germany

e-mail: [email protected]

H. Köhler, Dr. med. vet.

Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health,

Naumburger Str. 96a, 07743 Jena, Germany

e-mail: [email protected]

Corresponding author:

E. M. Liebler-Tenorio, Prof. Dr. med. vet.

Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health,

Naumburger Str. 96a, 07743 Jena, Germany

Tel +49 3641-8042411, Fax +49 3641-8042251

e-mail: [email protected]

(Manuscript submitted)

Page 64: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

60

4.1 Summary

Mycobacterium avium subsp. hominissuis (MAH) is an opportunistic pathogen and

causes infections in humans and animals. In this study, 18 goat kids were orally

inoculated with a high total dose of MAH. One group of goats (n=9) developed severe

clinical disease until 2-3 month after inoculation (mpi). At necropsy, ulcerative and

granulomatous lesions in organized gut-associated lymphoid tissue (oGALT) and

granulomas with extensive necrosis in intestinal lymph nodes (ILN) predominated.

Cultivation revealed MAH in all lesions and systemic spread. A second group of

goats were healthy at the end of the trial (13 mpi), however, all had extensive

granulomas in ILN but no extra-intestinal spread of bacteria. Moderate fecal shedding

occurred in all goats until 2 mpi. Further histological characterization of granulomas

revealed solid non-necrotic, necrotic, calcified and fibro-calcified granulomas with

striking resemblance to those seen in human and bovine tuberculosis. The two

different courses of disease with highly heterogenic lesions, systemic spread in goats

with severe clinical disease and the development of granulomas of all stages in the

surviving goats, makes the experimental infection of goats with MAH a valuable

model for TB research. This model might allow new insights into host-pathogen

interaction and anti-mycobacterial compound testing.

Keywords: communicable diseases, tuberculosis, nontuberculous mycobacteria,

Mycobacterium avium Complex, pathology, genotype, goat, disease models/animal

4.2 Introduction

Mycobacterium avium subsp. hominissuis (MAH) is an environmental opportunistic

pathogen, currently considered as potential zoonotic agent and was first recognized

as subspecies of Mycobacterium avium in 2002 (MIJS et al. 2002; TRAN u. HAN

2014). Mycobacterium avium is part of the M. avium complex (MAC) which belongs

to the large family of non-tuberculous mycobacteria (NTM) (BIET u. BOSCHIROLI

2014; ORME u. ORDWAY 2014). The incidence of NTM associated diseases is

increasing worldwide in developed countries with MAC as leading cause in humans,

while Mycobacterium tuberculosis complex (MTC) infections are declining (WEISS u.

Page 65: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

61

GLASSROTH 2012; BRODE et al. 2014). Epidemiological data are limited, because

NTM infections are predominantly not notifiable in humans and animals (WEISS u.

GLASSROTH 2012). In Queensland, Australia, a rise in incidence rates of NTM

infections in humans from 4.85/100.000 in 1999 to 5.7/100.000 in 2005 was reported,

while those for MTC infections were almost constant with 2.5/100.000 and

2.6/100.000, respectively (BRODE et al. 2014). Little is known about the prevalence

of NTM infections in animals with the exception of paratuberculosis, a chronic

granulomatous enteritis in ruminants, caused by Mycobacterium avium subsp.

paratuberculosis (MAP) (ELZE et al. 2013). Pigs were found to be particularly

susceptible for experimental and natural infections with MAH ( AGDESTEIN et al.

2012; AGDESTEIN et al. 2014). An epidemiological study of hunted wild boars in

Spain revealed a prevalence of 16.8 % NTM without further sub-classification for

MAH and 38.3 % MTC species (GARCÍA-JIMÉNEZ et al. 2015). A survey of

slaughtered pigs from Czech Republic that received peat as feed supplement,

revealed up to 75 % of mycobacterial isolates were MAH (MATLOVA et al. 2005). In

recent years, an increasing number of cases of MAH infections were reported in a

variety of animal species (GLAWISCHNIG et al. 2006; MÖBIUS et al. 2006; HAIST et

al. 2008; KRIZ et al. 2010; KLANG et al. 2014).

In pigs, MAH infections usually present as silent infections without clinical signs

mainly detected at slaughter. Granulomatous lesions of the digestive tract and

intestinal lymph nodes (ILN) predominate while disseminated disease is a rare

condition (AGDESTEIN et al. 2012; AGDESTEIN et al. 2014). Case reports in dogs,

horses, cat and red deer described severe protracted disease with fever, emaciation

and diarrhea. Granulomatous lesions of the digestive tract, ILN and systemic spread

were found accompanied by numerous acid fast bacilli (GLAWISCHNIG et al. 2006;

HAIST et al. 2008; KRIZ et al. 2010; KLANG et al. 2014). In humans, MAH causes

lymphadenitis in children and chronic lung disease in patients with cystic fibrosis,

chronic obstructive pulmonary disease and disseminated disease in patients with

acquired immunodeficiency syndrome (GRIFFITH et al. 2007; LAHIRI et al. 2014).

A current problem of screening tests for tuberculosis in animals and humans and

paratuberculosis in ruminants are cross reactions with M. avium subspecies causing

Page 66: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

62

false positive and false negative results (MANCUSO et al. 2012; BIET u.

BOSCHIROLI 2014). The initial aim of this study was the evaluation of specific

diagnostic antigens for paratuberculosis with an experimental infection of goats with

either MAP or MAH. Two to 3 month after inoculation (mpi) clinical signs progressing

to severe disease were observed unexpectedly in 50 % of the MAH infected goats.

The remaining MAH infected goats were healthy at necropsy 13 mpi. In the following,

the characteristics of the MAH strain used for inoculation, lesions and tissue

distribution of MAH in the infected goats and fecal shedding of MAH are reported.

This aims to raise awareness of possible infections of small ruminants with MAH and

of their similarities to MTC infections (DOBBERSTEIN u. WILMES 1937; NIEBERLE

1937; WANGOO et al. 2005; DEBI et al. 2014; PESCIAROLI et al. 2014; LENAERTS

et al. 2015)

4.3 Material and methods

Animals

This study was carried out in strict accordance with European and National Law for

the Care and Use of Animals. The protocol was approved by the Committee on the

Ethics of Animal Experiments and the Protection of Animals of the State of Thuringia,

Germany (Permit Number: 04-002/12). All experiments were done in containment of

biosafety level (BSL) 2 under supervision of the authorized institutional Agent for

Animal Protection. During the entire study, every effort was made to minimize

suffering.

Twenty-four goats of the breed “Thüringer Wald Ziege” (20 males, 4 females) from a

herd without history of paratuberculosis were used. Upon arrival at the animal

facilities at the age of 8-19 days, all goat kids were tested negative for MAH by fecal

culture. Infected and control animals were maintained in separate stables on straw

bedding. Kids were initially fed with milk replacer and subsequently with pelleted

concentrate for goats as described (KÖHLER et al. 2015). Water and hay were given

ad libitum. Male kids were castrated at the age of 6-8 weeks. Goats were treated

Page 67: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

63

against endo- and ectoparasites and received vitamin B supplementation (KRÜGER

et al. 2015).

Characterization and preparation of bacteria used for inoculation

MAH strain 09MA1289 used for inoculation was isolated from the enlarged lymph

node of a slaughtered pig in Southern Germany in 2009 using Stonebrink medium

with Pyruvat and PACT and Löwenstein-Jensen medium with Glycerol and PACT

(Bioservice Waldenburg). The isolate was maintained and propagated in Middlebrook

7H9 broth containing glycerine and 10% OADC, (MB, Becton Dickinson, Heidelberg,

Germany). Genomic DNA was prepared by the cetyltrimethylammonium bromide

method (VAN SOOLINGEN et al. 1991).

Subspecies identity of MAH strain 09MA1289 was verified by examination for the

presence of IS1245 (GUERRERO et al. 1995), absence of IS900 (ENGLUND et al.

1999), absence of IS901 (KUNZE et al. 1992) and presence of flanking region of

IS901 FR300 without the IS901 element (NISHIMORI et al. 1995). For genotyping,

restriction fragment length polymorphism (RFLP) analysis based on the insertion

sequence IS1245 (IS1245-RFLP) was conducted using the short probe of IS1245

(JOHANSEN et al. 2005) to prevent cross hybridization between IS1245 and IS1311

(MÖBIUS et al. 2006). Strain 09MA1289 was further characterized by mycobacterial

interspersed repetitive units-variable-number tandem repeat (MIRU-VNTR) typing

(RADOMSKI et al. 2010; FRITSCH et al. 2012).

As potential virulence markers, presence of ISMpa1 was determined by PCR using

the primers P2 and P3 (OLSEN et al. 2004) and presence of non-serovar-specific

glycopeptidolipid (nsGPL) genes (gtfA, rtfA, mtfC) and ser2 genes (mdhtA, merA,

mtfF) involved in the synthesis of GPLs in the cell wall of M. avium using published

primers (JOHANSEN et al. 2009).

The inoculum was prepared essentially as described elsewhere with modifications

(KÖHLER et al. 2015). Bacterial inoculum stocks were prepared from batch cultures

in MB. After centrifugation of the stocks, bacterial wet mass was adjusted to 10 mg

per dose using PBS. The bacterial inoculum of MAH strain 09MA1289 amounted to

0.78 - 4.1 x 109 cfu/dose.

Page 68: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

64

Study Design

Eighteen goats (No. 1-18) were inoculated starting at the age of 10-21 days, 10

times, every 2-4 days, with MAH mixed in milk replacer. The total inoculation dose

was 2.13 x 1010 cfu MAH per goat. Six control goats received milk replacer only.

Clinical signs were recorded daily and body weight every 4 weeks. Blood and fecal

samples were collected every 4 weeks also. Fecal samples were cultured for MAH.

Unexpectedly, 50% of goats infected with MAH had to be necropsied between 66-85

days post first inoculation (dpi) when clinical signs required euthanasia in 7 goats

because of animal welfare reasons. Two goats died spontaneously. The surviving

and control goats were euthanized and necropsied as scheduled between 362-392

dpi.

Cultural isolation of MAH from feces and tissues

Cultural isolation of MAH from feces and tissues was performed as described

recently (KÖHLER et al. 2015). Cultures were incubated up to 3 months at 37 ± 2 °C

and checked regularly for contamination and occurrence of visible colonies.

Presence or absence of colony growth was recorded.

Necropsy and sampling

At necropsy macroscopic lesions were documented and tissues sampled for

histology, immunohistochemistry (IHC) and cultivation of MAH as described

(KRÜGER et al. 2015). The following specimens were collected: duodenum, jejunum,

jejunal Peyer´s patches (JPP), ileal Peyer´s patch (IPP), organized gut associated

lymphoid tissue (oGALT) next to the ileocecal valve (ICVPP) and in the proximal

colon (PCPP), cecum, colon, rectum, intestinal lymph nodes (ILN) including proximal

and distal jejunal (J-LN), ileocolic (ICV-LN) and colonic lymph nodes (Co-LN). A

thorough necropsy with collection of specimens was performed immediately

subsequently as described elsewhere (KRÜGER et al. 2015).

Page 69: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

65

Histology and IHC

Extensive mineralized ILN were decalcified with HCL-EDTA decalcifying solution

(Richard Allen Scientific, Microm International, Germany) for 12 h prior to tissue

embedding. Lesions were examined in hematoxylin and eosin (HE)-stained formalin-

fixed paraffin embedded (FFPE) sections. Granulomatous infiltrates and granulomas

were graded from mild to severe. Staging of granulomas was adapted from a

previous study (WANGOO et al. 2005) (Tab. 4.1). MAH and mycobacterial material

were labeled in FFPE sections of all intestinal sites, ILN, liver, lung, tonsil, hepatic

LN, mediastinal LN, superficial inguinal LN and tissues with granulomas by IHC

(KRÜGER et al. 2015). Sections of an IPP with many MAH were used as positive

controls. As negative control the primary antibody was replaced by a polyclonal

antiserum against unrelated antigen. Negative controls of consecutive sections were

included for all tissues with necrosis and mineralization to verify specificity. MAH and

mycobacterial material were quantified according to an adapted scoring scheme

(KRÜGER et al. 2015) (Tab. 4.2).

Page 70: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

66

Table 4.1 Characterization of granulomas in oGALT and lymph nodes of goats

infected with MAH

Granuloma stage

Cellular composition Necrosis and mineralization

Fibrosis

1 (Initial)

epitheloid macrophage cluster, ± MGCs, interspersed and encircling lymphocytes,

single neutrophils

no necrosis -

2 (Solid) epitheloid macrophages, ± MGCs, interspersed with

lymphocytes and neutrophils

minimal necrosis and/or

mineralization

incomplete,

thin: -/+

3 (monocentric necrosis and

mineralization)

± epitheloid macrophages, ± MGCs, lymphocytes and

neutrophils, surround necrosis and/or mineralization

monocentric caseous necrosis

and/or mineralization

variable,

- / +++

4 (multicentric necrosis and

mineralization)

± epitheloid macrophages, ± MGCs, lymphocytes and

neutrophils, surround necrosis and/or mineralization

multicentric caseous necrosis

and/or mineralization

variable,

- / +++

5 (fibrotic organization)

minimal cellular infiltrate

fibrosis > necrosis and mineralization

+++

oGALT, organized gut-associated lymphoid tissue; MGCs, multinucleated giant cells;

-, not present; +, mild; ++, moderate; +++, severe; ±, missing or present at variable

number.

Page 71: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

67

Table 4.2 Scoring of labeled MAH and mycobacterial material in tissue sections by

IHC

Quantity Distribution

None < 2 MAH per section, no labelling in n and m

Single <20 % EM/ MGC with single MAH and/or single labeling of mycobacterial material in n+m

Few 20-50 % EM/MGC with 1-10 MAH and/or predominantly single to moderate labeling of mycobacterial material in n+m

Many 50-75 % EM/MGC with >10 MAH and < 50% with uncountable MAH and/or predominantly moderate to extensive labeling of mycobacterial

material in n+m

Numerous >75 % EM/MGC with >10 MAH and > 50% with uncountable MAH and/or predominantly extensive labeling of mycobacterial material in

n+m

MAH, Mycobacterium avium subsp. hominissuis; n, necrosis; m, mineralization; EM,

epitheloid macrophages; MGC, multinucleated giant cells; adapted from Krüger et al.

4.4 Results

Characteristics of MAH

Isolate 09MA1289 used as inoculum was verified as MAH based on presence of

IS1245 and FR300 [311 bp] and absence of IS901 and IS900. It exhibited the same

MIRU-VNTR profile 22221129 corresponding to INMV 46 as detected previously in a

porcine MAH isolate from France (RADOMSKI et al. 2010). IS1245-RFLP resulted in

a 12-banded pattern (Fig. 4.1). Strain 09MA1289 harbored ISMpa1 and was positive

for all nsGPL and ser2 genes examined (gtfA, rtfA, mtfC, mdhtA, merA, and mtfF).

(A) Goats with severe disease (necropsy at 66-85 dpi)

Clinical signs.

Nine of 18 goats inoculated with MAH developed elevated body temperature

immediately after inoculation and decreased food intake with mild depression at 40

dpi. Clinical signs progressed in severity until necropsy at 66–85 dpi and resulted in

Page 72: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

68

severely reduced body condition. Two goats died spontaneously; seven had to be

euthanized for animal welfare reasons.

MAH was culturally isolated in moderate amounts from the feces of 9/9 goats at the

end of the 4 week inoculation period and from 6/9 goats at 7-10 weeks post first

inoculation (wpi) and 1/1 goat at 11 wpi (data not shown).

Macroscopic, histologic and IHC findings.

All 9 goats were in bad body condition with serous atrophy of coronary and renal

adipose tissue, subcutaneous edema and transudate in body cavities. Lesions

predominated in the intestinal tract and regional lymph nodes.

Intestinal tract. Macroscopic lesions characterized by nodular thickening and

ulcerations were seen in JPP, IPP, ICVPP and PCPP with exception of the PCPP in

one goat (Tab. 4.3). The number of JPP affected varied from a few, at random, to all.

Ulcerations were especially extensive in IPP covering almost the complete length in 8

goats and thus resulting in ulcerated areas of up to 3 x 150 cm (Fig. 4.2A). Merely

small ulcerations with thickened mucosa (Fig. 4.2E) and multiple small, 1-mm foci of

mineralization were seen in goat no. 9. Ulcerations in ICVPP and PCPP were

multifocal and 2-10 mm in diameter. Multiple, round, 1-5 mm in diameter erosions

and ulcera occurred in cecum and colon outside oGALT in 6 goats. Chronic fibrous

serositis was frequently associated with altered oGALT.

Histologically, ulcerations in oGALT were always associated with granulomatous

infiltrates of variable severity and severe loss of the preexisting lymphatic tissue (Tab.

4.3). There were no granulomas. In 8 goats, granulomatous infiltrates in the

submucosa and rarely small foci in the overlying mucosa were composed of small

multifocal clusters of epitheloid cells, many lymphocytes, variable numbers of

macrophages, and many neutrophils (Fig. 4.2B). Foci of necrosis and mineralization

were regularly seen. Extensive infiltrates of epitheloid cells were found under intact

mucosa or superficial ulcerations, while deep ulcerations were associated with few

epitheloid cells (Fig. 4.2C) and granulation tissue. Single mycobacteria were present

in epitheloid cells and small amounts of mycobacterial material in areas with necrosis

and mineralization (Tab. 4.3, Fig. 4.2D).

Page 73: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

69

Lesions were different in one goat (no. 9). Diffuse granulomatous infiltrates

composed of many epitheloid cells, few macrophages, moderate numbers of

lymphocytes, many neutrophils, variable numbers of Mott cells and circumscribed

areas of necrosis and mineralization were present both in submucosa and mucosa

(Fig. 4.2F, G). More mycobacteria and mycobacterial material were found compared

to the other 8 goats (Tab. 4.3, Fig. 4.2H).

In all goats, granulomatous thrombi and emboli were frequent in submucosal and

subserosal lymphatics of IPP and occasionally in JPP, ICVPP and PCPP.

Granulomatous vasculitis of subserosal arterioles was observed occasionally. Mild to

moderate subserosal edema and serositis was frequent in sites with ulcerations.

Outside oGALT, small granulomatous infiltrates were seen in the mucosa of

duodenum, jejunum and cecum of 4 goats. Ulcerations in the large intestine outside

oGALT were not associated with granulomatous infiltrates, but presented with diffuse

infiltration of neutrophils.

Intestinal lymph nodes. Macroscopic lesions occurred in J-LN and ICV-LN of all

goats, but in the Co-LN of 3 goats only. Distal J-LN were more severely affected than

proximal J-LN. Lymph nodes were enlarged with multifocal to confluent necrosis and

mineralization of the cortex (Fig. 4.2I).

Histological lesions were dominated by granulomas with extensive mono- and

multicentric caseous necrosis, variable amount of mineralization and small islands of

preexisting lymphatic tissue (Fig. 4.2J). Areas of necrosis were irregularly surrounded

by few epitheloid cells, macrophages, lymphocytes and neutrophils. Formation of

collagenous capsules in the periphery of granulomas was absent, except adjacent to

the capsule of the lymph node (Fig. 4.2J). These features are compatible with stage

3 and 4 granulomas (Tab. 4.4). In addition, there were multifocal to confluent

infiltrates of epitheloid cells (Tab. 4.4). Single MAH were labeled in epitheloid cells

and mycobacterial material was found in variable quantity in areas with necrosis and

mineralization (Tab. 4.4, Fig. 4.2K).

In goat no. 9, confluent granulomatous infiltrates dominated. Stage 3 and 4

granulomas were present in lower numbers with less caseous necrosis and more

extensive mineralization than in the other goats (Tab. 4.4). Numerous mycobacteria

Page 74: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

70

were seen in epitheloid cells and there was extensive labeling for mycobacterial

material (Tab. 4.4).

Other organs. Few, small granulomatous infiltrates were regularly found in hepatic-

LN, retropharyngeal LN and mediastinal LN and rarely in tonsils and liver. A few

small stage 2 and 3 granulomas occurred in liver and mediastinal LN. In 2 goats,

there was a moderate to extensive granulomatous bronchopneumonia with

neutrophils and few giant cells. Single mycobacteria were detected in epitheloid cells

in one of these goats (no. 1). All goats had moderate to severe thymic atrophy and

multiple small periportal lympho-histiocytic infiltrates. Multifocal areas of renal tubular

necrosis were seen in 6 goats and focal catarrhalic bronchopneumonia in one.

Multiple fibrin thrombi were seen in kidneys, livers and lungs of 3 goats.

Cultural Isolation of MAH from tissues.

MAH was recovered in moderate to high amounts from all locations of oGALT and

intestinal lymph nodes of all necropsied goats (Tab. 4.3, 4.4). Cultural isolation of

MAH was also possible from extra-intestinal sites, most often from hepatic LN,

mediastinal LN and spleen, followed by retropharyngeal LN, liver and tonsil (data not

shown).

(B) Goats with mild disease / surviving group (necropsy at 365-390 dpi)

Clinical signs.

The 9 surviving goats developed a transient increase of body temperature and mild

depression until 60 dpi. Thereafter they were clinically healthy until the end of the

trial. MAH was culturally isolated in moderate to very high amounts from the feces of

these goats at the end of the four-week inoculation period and from 4/10 goats at 7-

10 wpi. Fecal shedding was no longer recorded from 11 wpi until the end of the

experiment.

Page 75: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

71

Macroscopic, histological and IHC findings.

All goats were in very good nutritional condition. Lesions were detected in all goats

predominantly in oGALT and ILN.

Intestinal tract. Multiple small granulomas were seen in IPP of 7 goats and single

small granulomas in some JPP of 2 goats (Fig. 4.3A). Some JPP were atrophic in 3

goats. Occasionally, mild serositis was associated with altered Peyer´s patches.

Histologically, granulomas of varying size and stage were seen in the submucosa of

JPP and IPP (Tab. 4.3). Monocentric stage 3 granulomas (Fig. 4.3B) predominated

with extensive central necrosis and mineralization surrounded by a layer of

granulomatous infiltrate with many epitheloid cells, few multinucleated giant cells

(MGC) and few lymphocytes followed by a layer of lymphocytes (Fig. 4.3C). In

addition, there were focal to multifocal granulomatous infiltrates in the mucosa and

submucosa of 3 goats (Tab. 4.3). Granulomatous lymphangitis, but no granulomas

were found in the subserosa of the IPP in one goat. Mycobacteria were detected

rarely in granulomas in the IPP: single mycobacteria in epitheloid cells and MGCs

(Fig. 4.3D) of 1 goat and small amounts of mycobacterial material in necrosis and

mineralization of 3 goats (Tab. 4.3).

Segmental to diffuse atrophy affected lymphoid follicles in some JPP of all, ICVPP of

2 and PCPP of 3 goats, while age-dependent atrophy was observed in IPP. Mild

filamentous serositis was frequently associated with atrophic Peyer`s patches.

Intestinal lymph nodes. Extensive granulomas in severe cases completely replacing

the lymph node were seen in many J-LN and ICV-LN in all goats (Tab. 4.4, Fig.

4.3E).

Histologically, extensive granulomas of stage 4 and 5 dominated, while

granulomatous infiltrates and stage 2 and 3 granulomas were occasionally present in

J-LN and ICV-LN (Tab. 4.4). Granulomas of stage 3 and 4 had fibrotic capsules of

varying thickness and only minimal infiltrates of epithelial cells and MGC´s. Fibrotic

organization predominated in some granulomas (stage 5 granulomas, Fig. 4.3F, G).

Granulomatous lymphangitis was seen in the capsule of the lymph nodes in 2 goats.

Co-LN had few small granulomas of stage 3 and 4 and focal granulomatous infiltrates

(Tab. 4.4). Single MAH and small amounts of mycobacterial material were labeled in

Page 76: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

72

the necrosis and mineralization of some granulomas (Fig. 4.3H, Tab. 4.4). Single

mycobacteria were found in epitheloid macrophages and MGCs of 2 goats.

Other organs. Few small stage 2 and 3 granulomas and granulomatous infiltrates

were present in liver LN and retropharyngeal LN of one goat. Infiltrates of few

epitheloid cells or single MGCs were associated with bronchus associated lymphatic

tissue of two goats as well as pulmonary interstitium and renal cortex of one goat

each. Multifocal granulomatous infiltrates were observed in the mediastinal LN of one

goat.

Most goats had multiple small periportal lympho-histiocytic infiltrates. Parasite-

induced alterations were observed as filamentous peritonitis at forestomachs and

diaphragm and eosinophilic granulomas and infiltrates in tonsils, R-LN and kidneys.

Cultural isolation.

MAH were isolated from few sites of oGALT and ILN, but not from extra-intestinal

sites (Tab. 4.3, 4.4).

(C) Control animals (necropsy at 362-392 dpi)

Control animals did not developed clinical signs throughout the trial. Neither

macroscopic nor histological lesions were seen. Mycobacteria were not detected by

IHC or culture.

Page 77: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

73

Table 4.3 Macroscopic and histological lesions with detection and cultivation of MAH

in oGALT of experimentally infected goats

No

JPP

IPP

ICVPP

PCPP

M

micro. lesion

M

micro. lesion

M

micro. lesion

M

micro. lesion

gI Gr MAH

gI Gr MAH

gI Gr MAH

gI Gr MAH

I C I C I C I C

66-85 dpi 1 N/U + - a +

N/U + - a +

N ++ - - +

N + - - + 2

† N/U ++ - a + N/U + - a + N/U ++ - a + N/U ++ - - +

3 N/U + - a + N/U +++ - a + N/U + - - + N/U ++ - - + 4

† N/U ++ - a + N/U ++ - a + N/U ++ - - + N/U + - - +

5 N/U +++ - a + N/U ++ - a + N/U + - - + N/U ++ - a + 6 N/U ++ - a + N/U + - a + N/U nd nd nd N/U + - a + 7 N/U ++ - a + N/U ++ - a + N/U ++ - - +

N/U + - - +

8 N/U ++ - a + N/U +++ - a + N/U ++ - a + - - - - + 9 N/U +++ - c + N/U +++ - c + N/U +++ - b + N/U ++ - b +

365-390 dpi 10 A - - - -

- - 3 - -

- - - - -

A - - - - 11 A + 1,3

* - - N + 2,3,4 - - - - - - - - - - - -

12 N - 3 - - N - 2,3* - - - - - - - - - - - -

13 - - 3 - - - ++‡ - - + - - - - - - - - - -

14 N - 4 - - N - 2,3* a - - - - - - - - - - -

15 A - - - - N - 2,3* a - N - - - - - - - - -

16 - + 1 - - N ++ 1,3* - + - - - - - - - - - -

17 - + 1,2* - - N - 3 a + - - - - - - - - - -

18 - - 3 - - N - 3 - - - - - - - - - - - -

MAH, Mycobacterium avium subsp. hominissuis; oGALT, organized gut-associated lymphoid tissue,

dpi, days post first inoculation; JPP, jejunal Peyer´s patch; IPP, ileal Peyer´s patch; ICVPP, oGALT in

the colon next to the ileocecal valve; PCPP, oGALT at the end of the proximal colon; No, animal

number; M, macroscopic lesion; micro., microscopic; gI, granulomatous infiltrate; Gr, granuloma

stages; I, immunohistochemistry; C, bacterial culture; N, nodular thickened; U, ulceration; A, atrophy;

+, mild or positive in bacterial culture; ++, moderate; +++,severe; a, single; b, few; c, many; d,

numerous; -, no gross lesion/ microscopic lesions/ <2 or no mycobacteria by immunohistochemistry/no

mycobacteria in bacterial culture; nd, not done.

*Predominant granuloma stage.

†Spontaneously deceased animal.

‡Granulomatous lymphangitis.

Page 78: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

74

Table 4.4 Macroscopic and histological lesions with detection and cultivation of MAH

in ILN of experimentally infected goats

No

J-LN proximal J-LN distal

ICV-LN

Co-LN

M

micro. lesion

M

micro. lesion

M

micro. lesion

M

micro lesion

gI Gr MAH

gI Gr MAH

gI Gr MAH

gI Gr MAH

I C I C I C I

66-85 dpi 1 X ++ 3,4

* a + X ++ 3,4

* a +

X ++ 3*,4 a +

- + 3 - 2

† X ++ 3,4

* a + X ++ 2,3,4

* c + X ++ 3,4

* b + X ++ 3,4

* -

3 X +++ 3,4* a + X ++ 2,3,4

* b + X ++ 3,4

* a + - + - -

4† X ++ 3,4

* a + X ++ 3,4

* a + X ++ 3

*,4 a + - ++ - -

5 X +++ 3*,4 a + X +++ 3,4

* b + X ++ 3,4

* a + - - - -

6 X +++ 3 a + X +++ 3,4* c + X ++ 3,4

* c + X ++ 3

*,4 b

7 X ++ 3,4* b + X ++ 3,4

* c + X ++ 3,4

* c + X ++ 3,4

* a

8 X +++ 3,4* b + X +++ 3,4

* b + X +++ 3

*,4 c + - - - -

9 X +++ 2,3,4* d + X +++ 3,4

* d + X +++ 3,4

* d + - + - a

365-390 dpi 10 X ++ 2,3,4,5

* a - X ++ 3,4

*,5 a -

X - 3 a -

- - 3*,4 -

11 X + 3,4*,5 a + X - 4 a - X + 3,4

* a - - - - -

12 - - 3*,4 a + X + 3,5

* a - - - 3 a - - - 2 -

13 X - 4 a + X ++‡ 3,4

* a + X ++

§ 2,4

*,5 a + - - - -

14 X - 3,4* a - X +

‡ 3,4

* a - X - 2,3,4

* a - - + 3 -

15 X + 3*,4,5 a - X ++ 3,4,5

* a - X - 3

*,4 a - - - - -

16 X ++ 2,3,4* a - X ++ 2,3,4

* a - X + 3 a - - - - -

17 X - 3,4* a - X - 3,4

* a - X - 3,4

* a - - - - -

18 X ++ 3,5* a - X + 3,4

* a + X - 4 a - - - - -

MAH, Mycobacterium avium subsp. hominissuis; ILN, intestinal lymph node, dpi, days post first

infection; J-LN, jejunal lymph node; ICV-LN, ileocolic lymph node; Co-LN, colonic lymph node; No,

animal number; M, macroscopic lesion; micro., microscopic; gI, granulomatous infiltrate; Gr,

granuloma stages; I, immunohistochemistry; C, bacterial culture; X, present; +, mild or positive in

bacterial culture; ++, moderate; +++,severe; a, single; b, few; c, many; d, numerous, -, no gross

lesion/microscopic lesions/ <2 or no mycobacteria by immunohistochemistry/no mycobacteria in

bacterial culture.

*Predominant granuloma stage.

†Spontaneously deceased animal.

‡Granulomatous lymphangitis.

Page 79: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

75

Figure 4.1 IS1245-RFLP patterns of PvuII digested genomic DNA from German M.

avium strains.

Lanes 1 and 2: MAA strains from deer and goose, Lane 3: MAS type strain DSM

44175, Lane 4: MAH bovine strain 00A0920, Lane 5: MAH porcine strain 09MA1289.

Lanes M: molecular weight (Mw) markers. MAA: M. avium subsp. avium, MAS: M.

avium subsp. silvaticum, MAH: M. avium subsp. hominissuis.

Page 80: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

76

Figure 4.2

Gross and microscopic lesions with immunohistochemical detection of MAH in JPP,

IPP and ILN of goats 66-85 dpi.

A. Diffuse ulceration of the IPP; goat No. 3.

B. Moderate amount of granulomatous infiltrate underneath extensive ulceration (U)

and small foci of granulomatous infiltrates in the lamina propria (arrow) of a JPP;

goat No. 7.

C. Granulomatous infiltrate is composed of many lymphocytes and neutrophils

(arrowhead), but only single epitheloid cells (arrows); higher magnification of 2B.

D. Single MAH in an epitheloid cell (arrow); consecutive section of 2C.

E. Small circumscribed ulcerations in the IPP (arrows); goat No. 9.

F. Extensive granulomatous infiltrate in the submucosa and lamina propria with

circumscribed necrosis and mineralization (left lower corner) and limited mucosal

ulceration (U) of the IPP; goat No. 9.

G. Granulomatous infiltrate is composed of many epitheloid cells, few lymphocytes

and neutrophils (arrowhead); higher magnification of 2F.

H. Many MAH in epitheloid cells; consecutive section of 2G.

I. Severely enlarged distal jejunal LN with cortical necrosis and mineralization with

radial appearance; goat No. 7.

J. Cortex of proximal jejunal LN replaced by multicentric granulomas (stage 4) with

extensive caseous necrosis (N), multiple foci of mineralization (M), an island of

preexisting lymphoid tissue (L) and moderately thickened lymph node capsule.

K. Large amount of fine-grained to clustered mycobacterial material in an area of

necrosis and mineralization, area delineated in 2J.

Page 81: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

77

Hematoxylin and eosin staining in panels B, C, F, G, J; IHC in panels D, H, K; scale

bar = 1 cm in panels A, E, I; scale bar = 500 µm in panels B, F, J; scale bar = 100 µm

in panels C, G, K; scale bar = 20 µm in panels D, H

Page 82: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

78

Figure 4.3

Gross and microscopic lesions with immunohistochemical detection of MAH in IPP

and ILN of goats 365-390 dpi.

A. Thickened, nodular IPP; goat No. 16.

B. Granuloma (stage 3) with central necrosis (N) and mineralization (arrows)

surrounded by granulomatous infiltrate (gI) and a layer of lymphocytes (L);

microscopic view of 3A.

C. Granulomatous infiltrate is composed of epitheloid cells, multinucleated giant cells,

lymphocytes and neutrophils. The layer of lymphocytes is visible at top. Area

delineated in 3B.

D. Single MAH (arrow) in a multinucleated giant cell; consecutive section of 3C.

E. Enlarged and completely mineralized distal jejunal LN with a thickened capsule;

goat No. 12.

F. Multicentric granuloma (stage 5) with prominent fibrotic organization (f) and

multiple areas of mineralization (M).

G. Fibrosis and mineralization without granulomatous infiltrate, area delineated in 3F.

H. Single MAH (arrow) in an area of mineralization, consecutive section of 3G.

Page 83: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

79

Hematoxylin and eosin staining in panels B, C, F, G; IHC in panels D, H; Panel F, G,

H decalcified; scale bar = 1 cm in panels A, E; scale bar = 500 µm in panels B, F;

scale bar = 100 µm in panels C, G; scale bar = 20 µm in panels D, H

Page 84: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

80

4.5 Discussion

Oral inoculation of goats with MAH resulted in infection of all animals which indicates

that goats are highly susceptible adding them to the increasing list of species in

which MAH infections have been found (GLAWISCHNIG et al. 2006; MÖBIUS et al.

2006; HAIST et al. 2008; KRIZ et al. 2010; AGDESTEIN et al. 2012; AGDESTEIN et

al. 2014; KLANG et al. 2014). The MAH strain used for inoculation was a porcine

isolate with a MIRU-VNTR genotype unique for Germany until now. The presence of

all selected GPL synthesis genes and ISMpa1 distinguishes this MAH strain from

those commonly isolated in Norway (JOHANSEN et al. 2009), but is common in MAH

strains from Germany (P. Möbius, pers. comm.). Although no further data concerning

host-pathogen interaction are available, there is no indication for an increased

pathogenicity of this particular strain. The high inoculation dose might be the cause

for the severe course of the disease in part of the animals.

Two distinct courses of infection were seen in the inoculated goats: (a) severe, fatal

disease and (b) transient mild infection with persisting lesions. Since all goats

received the same dose of MAH, were kept under identical conditions and had similar

age, key factors for the outcome of the infection are most likely differences in the

individual host’s immune responses. As goats are an outbred species, genetic

heterogeneity has to be expected although all animals came from one herd and one

breed. Comparable observations were made after infection of goats with MAP in

previous trials (KÖHLER et al. 2015; KRÜGER et al. 2015). This turns the goat into

an appropriate animal model, since inter-individual differences naturally occur in TB

in humans and cattle, and the currently used animal models are not able to establish

this heterogeneity (VAN RHIJN et al. 2008).

The closer analysis of the lesions in the goats with severe disease allows insights in

the pathogenesis of the infection. Based on the distribution of lesions, oGALT is the

predominant port of entry as reported for the closely related MAP, although the

invasion via enterocytes as seen in mice cannot be excluded (MOMOTANI et al.

1988; SANGARI et al. 2001; SIGURDARDÓTTIR et al. 2001). The marked infiltration

of lymphocytes and low number of epitheloid cells are indications for an intense local

immune response. This appears to limit proliferation of MAH, but on the other hand

Page 85: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

81

results in extensive tissue destruction, e.g. ulcerations of the mucosa overlying

oGALT and necrosis in the draining lymph nodes. Intestinal ulcerations and acute

clinical signs are not commonly associated with mycobacterial infections in goat kids,

but should be included in the differential diagnosis of ulcerative conditions (UZAL u.

SONGER 2008). Distribution of lesions and type of inflammatory infiltrate closely

resemble those described in case reports of MAH infections in various species

(GLAWISCHNIG et al. 2006; HAIST et al. 2008; KRIZ et al. 2010; KLANG et al.

2014). They also have close resemblance to lesions and their distribution described

in bovine tuberculosis when calves were orally inoculated with Mycobacterium bovis

(DOBBERSTEIN u. WILMES 1937; NIEBERLE 1937). Preferential localization of

ulcerations and granulomas is described in the ileocecal region in human intestinal

tuberculosis (DEBI et al. 2014). The herein described systemic spread of MAH

infection and rapid disease progression in the young goats has also similarity to

human pediatric TB and might represent a model for TB infection in this specific age

group (MARAIS et al. 2004).

Clinically mild infection with persisting lesions is common in natural and experimental

MAH infection of pigs (AGDESTEIN et al. 2012; AGDESTEIN et al. 2014). In the

surviving goats, lesions were seen at the same sites as in the pigs and as in the

goats with severe disease. This indicates the ability of the host`s immune system to

limit the inflammation after entry by forming granulomas. Because of the macroscopic

resemblance of granulomas, MAH should be considered as differential diagnosis in

field cases of intestinal TB in ruminants (DOBBERSTEIN u. WILMES 1937;

NIEBERLE 1937; PESCIAROLI et al. 2014).

Granulomas, solid without necrosis, with necrosis and mineralization, with fibrotic

organization, are major hallmarks of TB besides cavitary lesions in the lung

(BASARABA 2008; LENAERTS et al. 2015). The goats in this study developed a

wide spectrum of morphologically different granulomas. Because of the great

resemblance to granulomas observed in cases of human and bovine TB (WANGOO

et al. 2005; LENAERTS et al. 2015), MAH infections of goats may serve as a

comparative model for TB research. In contrast to goats, mice, the predominant

species used in experimental TB research, lack those key features, but develop

Page 86: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

82

poorly organized granulomas, high bacterial burden and finally die after protracted

infection (BARRY et al. 2009; LENAERTS et al. 2015). Cavitary lesions were not

seen in this study, however this is reported in natural and experimental TB infections

of goats developing cavitary lesions and thus are one of few species that exactly

model this feature of TB (SANCHEZ et al. 2011; LENAERTS et al. 2015).

The range of morphologically different granuloma stages that were induced in this

model could be utilized for testing of anti-mycobacterial compounds.

Pharmacodynamics and pharmacokinetics of drugs are directly influenced by certain

characteristics of granulomas like necrosis, mineralization and extensive fibrosis

(BASARABA 2008; BARRY et al. 2009). In addition, the relative small size of goats

would allow the utilization of state of the art live imaging systems to evaluate

development of lesions under drug therapy (DE VAL et al. 2011; LENAERTS et al.

2015).

Finally, MAH infections of goats may also be of relevance for public health. Fecal

shedding seen in all goats until 2 mpi may cause contamination of the environment

and might pose a health risk to immunocompromised individuals, children and the

elderly (HAIST et al. 2008; KRIZ et al. 2010). Especially children may come in close

contact to goats in zoos and petting farms (STIRLING et al. 2008). If infected animals

are slaughtered, contamination of the slaughterhouse and meat products is

conceivable as indicated by detection of DNA from MAH by PCR in retail meat from

cattle and pigs (KLANICOVA et al. 2011; AGDESTEIN et al. 2014; KLANICOVA-

ZALEWSKA u. SLANA 2014).

In this study, oral inoculation of goats with MAH induced infections in 100% of

animals, with two different courses of clinically overt and silent disease and a variety

of morphological different lesions that resemble different aspects of TB pathology.

Different inoculation modes and dose titration might help to adopt the herein

described experimental goat model to specific research objectives. Overall this

approach could provide new insights into progression of mycobacterial diseases and

anti-mycobacterial compound testing and overcome obstacles that occur in TB

research with currently used animal models.

Page 87: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 1

83

Conflict of Interest Statement

The authors declare they have no conflicts in relation to this work.

Authors´ contributions

HK and ELT designed the study. PM performed identification and characterization of

the MAH strain used for inoculation. HK performed the inoculation of goats and the

collection of samples for bacteriologic examination. JS and ELT performed the gross

pathological examination and tissue collection for histology. JS performed the

histopathological and immunohistochemical investigations, and drafted the

manuscript. ELT supervised the study. HK, PM and ELT helped in critical discussion

of the results and revised the draft manuscript. All authors read and approved the

final manuscript.

Acknowledgement

This project was financially supported by the EMIDA-ERA-NET project

“Mycobactdiagnosis” (grant no. 031A083). The authors thank U. Brommer, S. Lied,

K. Steger, S. Werner, L. Wirker and L. Wolf for excellent technical support, the staff

of the animal facility of the FLI Jena and S. Fischer for dedicated care of the animals

and support with necropsies and W. Maginot for excellent photography. We also

thank S. Fischer and P. Reinhold for fruitful discussions.

References (Chapter 9)

Page 88: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

84

5 Manuscript 2

Characterization of tuberculous granulomas in different stages of progression

and associated tertiary lymphoid tissue in goats experimentally infected with

Mycobacterium avium subsp. hominissuis

Authors: Jan Schinköthe, Heike Köhler, Elisabeth M. Liebler-Tenorio

Affiliations:

Jan Schinköthe, DVM

Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health,

Naumburger Str. 96a, 07743 Jena, Germany

H. Köhler, Dr. med. vet.

Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health,

Naumburger Str. 96a, 07743 Jena, Germany

Corresponding author:

E. M. Liebler-Tenorio, Prof. Dr. med. vet.

Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health,

Naumburger Str. 96a, 07743 Jena, Germany

Tel +49 3641-8042411, Fax +49 3641-8042251

[email protected]

(Manuscript submitted)

Page 89: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

85

5.1 Summary

Oral infection of goats with Mycobacterium avium subsp. hominissuis (MAH) resulted

in a large variety of granulomas in organized gut-associated lymphatic tissues and

intestinal lymph nodes. To characterize the cellular composition of granulomas,

CD4+, CD8+, γδ, B lymphocytes and plasma, CD25+, CD68+, MHC-II+, Ki67+ and

endothelial cells were labeled in consecutive frozen sections by

immunohistochemistry and acid fast bacilli (AFB) by Kinyoun stain. Granulomas with

extensive necrosis, little mineralization and variable numbers of AFB surrounded by

many CD4+ T cells, but only few epitheloid macrophages were observed in severely

sick goats at 2-3 mpi. They were interpreted as exuberant immune reaction.

Organized granulomas with very few AFB were seen in clinically healthy goats at 13

mpi. The necrotic cores were surrounded by a zone of granulomatous infiltrate with

many epitheloid macrophages and few lymphocytes. This zone was initially wide and

highly vascularized and became progressively smaller. It was enclosed by an

increasing layer of connective tissue. All organized granulomas were surrounded by

compartimentalized tertiary lymphoid tissue. The granulomas in experimental

infection of goats with MAH reflect the heterogeneity of lesions seen in mycobacterial

infections of humans and ruminants and are therefore valuable for comparative

research.

Keywords: granuloma, immune response, animal model, Mycobacterium avium

subsp. hominissuis, tuberculosis, lymphoid tissue

5.2 Introduction

A hallmark of Mycobacterium (M.) tuberculosis complex (MTC) infection is the

presence of structured granulomas with central necrotic cores with variable

mineralization surrounded by epitheloid macrophages, multinucleated giant cells

(MGC), other inflammatory cells and connective tissue (WANGOO et al. 2005;

ULRICHS u. KAUFMANN 2006; BASARABA 2008; RAMAKRISHNAN 2012). These

granulomas are sites of host-pathogen interactions that on the one hand may limit

mycobacterial growth and tissue pathology, but on the other hand can be exploited

Page 90: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

86

by the mycobacteria for survival (EHLERS u. SCHAIBLE 2012; RAMAKRISHNAN

2012). They are highly dynamic structures with an enormous morphological

heterogeneity within an individual and even within one organ (EHLERS u. SCHAIBLE

2012; LENAERTS et al. 2015). Thus, they are important read out parameters for the

stage of infection and the reaction to anti-tuberculous treatments and animal models

that develop the spectrum of granulomas seen in human tuberculosis (TB) are highly

desirable.

The currently used small animal models in experimental TB research are often not

capable to reproduce the complete spectrum of granuloma heterogeneity (EHLERS

u. SCHAIBLE 2012; SHALER et al. 2013; LENAERTS et al. 2015). Most mouse

strains develop poorly organized granulomas. Granulomas with necrosis, but without

the important feature of calcification are described in C3HeB/FeJ mice (DRIVER et

al. 2012; LENAERTS et al. 2015). The guinea pig model overcomes these

shortcomings (LENAERTS et al. 2007; PALANISAMY et al. 2008). However, both

guinea pigs and mice develop only progressive fatal disease without a phase of

latency (BASARABA 2008; VAN RHIJN et al. 2008). Granulomas with morphology

comparable to human TB have been described in infected macaques; however,

ethical issues, costs and availability of animals are limitations of this model (VAN

RHIJN et al. 2008; LENAERTS et al. 2015). The zebrafish model revealed new

aspects of the interaction between M. marinum and the innate immune response due

to the unique opportunity of live imaging during granuloma formation, but zebrafish

embryos are not able to mount an adaptive immune response (DAVIS u.

RAMAKRISHNAN 2009; RAMAKRISHNAN 2012).

Cattle have been used in TB research for a long time, whereas goats have

increasingly gained acceptance in recent years. Both are naturally susceptible to M.

bovis and M. caprae, respectively, and may undergo a phase of latency (VAN RHIJN

et al. 2008; PESCIAROLI et al. 2014). Granulomas with a wide spectrum of

heterogeneity can be observed (WANGOO et al. 2005; VAN RHIJN et al. 2008;

SANCHEZ et al. 2011). They have been classified as: stage 1- clusters of epitheloid

macrophages, stage 2 - granulomas with minimal necrosis and variably thin fibrous

capsule, stage 3 – granulomas with central caseous and mineralized necrosis and

Page 91: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

87

complete fibrous capsule and stage 4 – granulomas with extensive multicentric

caseous and mineralized necrosis and marked peripheral fibrosis (WANGOO et al.

2005).

After experimental oral infection of goats with M. avium subsp. hominissuis (MAH),

an opportunistic, non- tuberculous mycobacterium, goats developed either severe

fatal disease with marked ulcerative and granulomatous lesions predominantly in

organized gut-associated lymphoid tissue (oGALT) and extensive caseonecrotic

granulomas in intestinal lymph nodes (ILN) at 2-3 month after inoculation (mpi) or a

wide variety of morphologically different granulomas in oGALT and ILN at 13 mpi.

The objectives of this investigation were to characterize the morphology of

granulomas seen in the goats infected with MAH and to compare them with those

seen in other granuloma models and in human TB. Thus cellular composition and

presence of MAH were examined in the different granulomas. A special focus was on

the layer of lymphocytes in the periphery of granulomas resembling tertiary lymphoid

tissue (3rd LT) described recently in human TB (ULRICHS et al. 2004; ULRICHS u.

KAUFMANN 2006), since it has been suggested that the interaction between

mycobacteria and 3rd LT determines the development of latency versus progression

in TB (ULRICHS et al. 2004; ULRICHS et al. 2005).

5.3 Material and methods

5.3.1 Tissue samples

The experimental infection of goat kids was carried out in strict accordance with

European and National Law for the Care and Use of Animals. The protocol was

approved by the Committee on the Ethics of Animal Experiments and the Protection

of Animals of the State of Thuringia, Germany (Permit Number: 04-002/12). All

experiments were performed in containment of biosafety level 2 under supervision of

the authorized institutional Agent for Animal Protection. During the entire study, every

effort was made to minimize suffering. A total of 24 goat kids of the breed “Thüringer

Wald Ziege” (20 males, 4 females) were used in this study as described recently

(Schinköthe et.al, submitted). In brief, 18 goat kids were orally inoculated 10 times

Page 92: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

88

every 2-4 days with a total dose of 2.13 x 1010 cfu MAH beginning at the age of 10-21

days. Between 2-3 mpi, 2 animals died spontaneously and 7 had to be euthanized for

animal welfare reasons because of severe clinical symptoms of elevated body

temperature, weight loss and apathy. The remaining 9 goats developed mild transient

increase in body temperature and depression around 2 mpi, recovered and remained

healthy until necropsied at 13 mpi.

Ulcerative lesions primarily restricted to the oGALT and granulomas in draining ILN

were seen in goats necropsied at 2-3 mpi and numerous granulomas of varying size

at the same sites in goats necropsied at 13 mpi. After macroscopic documentation of

lesions, multiple sites of altered oGALT and ILN were collected for histology,

immunohistochemistry (IHC) and bacterial cultivation. To investigate the cellular

composition, lesions and/or granulomas were collected from at least one jejunal

Peyer´s patch (JPP), ileal Peyer´s patch (IPP) and ILN of 5 of the 9 goats necropsied

at 2-3 mpi and from 1-2 JPP and ILN as well as 1-2 different sites of IPP in 9 out of

the 9 goats necropsied at 13 mpi. This resulted in a total of 6 JPP, 5 IPP, 5 ILN from

goats at 2-3 mpi and 13 JPP, 11 sites of IPP and 12 ILN from goats at 13 mpi.

These tissue samples were snap frozen in 2-methyl-butane at -70 °C immediately

after euthanasia. Consecutive frozen sections of 5 µm thickness were collected on

glass slides coated with chrome alum gelatin.

5.3.2 Staining of frozen tissue sections

Frozen sections were stained with hematoxylin and eosin (HE) for classification of

granulomas based on a previously described scheme (Tab. 5.1, Schinköthe et.al,

submitted.). The following 6 stages were differentiated: stage 1 (initial), stage 2

(solid), stage 3 (monocentric necrosis and mineralization), stage 4 (multicentric

necrosis and mineralization), stage 5 (fibrotic organization) and stage 6 (extensive

caseous necrosis). Azan staining was used to detect collagen fibers and the Kinyoun

stain to detect acid fast bacilli (AFB). (TB stain kit K, BD Diagnostics, US). Numbers

of AFB were semi-quantitatively assessed at x 1000 magnification in a reference

area (RA) of 2359 µm2 on representative sections (Tab. 5.3).

Page 93: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

89

Table 5.1 Characterization of granulomas in oGALT and lymph nodes of goats

infected with M. avium subsp. hominissuis

Granuloma stage

Cellular composition Necrosis and mineralization

Fibrosis

1 (Initial)

epitheloid macrophage cluster, ± MGCs, interspersed and

encircling lymphocytes, single neutrophils

no necrosis -

2 (Solid) epitheloid macrophages, ± MGCs, interspersed with

lymphocytes and neutrophils

minimal necrosis and/or

mineralization -

3 (Monocentric necrosis and

mineralization)

± epitheloid macrophages, ± MGCs, lymphocytes and

neutrophils surround necrosis and/or mineralization

monocentric caseous

necrosis and/or mineralization

variable,

- / +++

4 (Multicentric necrosis and

mineralization)

± epitheloid macrophages, ± MGCs, lymphocytes and

neutrophils surround necrosis and/or mineralization

multicentric caseous

necrosis and/or mineralization

variable,

- / +++

5 (Fibrotic organization)

minimal cellular infiltrate

fibrosis > necrosis and mineralization

+++

6 (Extensive caseous necrosis)

many lymphocytes, interspersed epitheloid macrophages and

neutrophils,

irregular necrosis with

minor mineralization

-

oGALT, organized gut-associated lymphoid tissue; MGCs, multinucleated giant cells;

±, missing or present at variable number ; -, not present; +++, severe;

5.3.3 Immunohistochemistry (IHC) for cell surface markers and proliferation

The cell surface markers CD4, CD8, TcR1-N24, CD79α, CD25, CD68, and MHC-II

were labeled in frozen sections. A monoclonal antibody against Ki-67 was used for

differentiation of proliferating cells and a polyclonal rabbit antiserum to human van

Willebrand Factor (vWF) for detection of endothelial cells. The monoclonal antibodies

used and cell types detected are summarized in table 5.2.

Frozen sections were air dried and fixed for 10 minutes (min) in acetone at room

temperature except for sections used for the detection of CD25 which were directly

transferred in 1:1 acetone-methanol at -20 °C for 10 min and for the detection of Ki-

67 which were fixed in neutral buffered formalin for 10 min at room temperature.

Page 94: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

90

Endogenous peroxidase was blocked by incubation with 0.06 % phenylhydrazine in

PBS at 37 °C for 40 min. Ki-67 antigen was retrieved by heating in citrate buffer in a

microwave oven for 10 min. Peroxidase-conjugated sheep anti-mouse IgG (NA931V,

GE Healthcare Europe, Freiburg, Germany) was used as second antibody for the

detection of CD8, TcR1-N24, CD68, and MHC-II antigen. The signals for CD25 and

CD79α were enhanced by incubation with biotinylated goat anti-mouse IgG (GE

Healthcare Europe GmbH, Freiburg, Germany ) and avidin-biotin-complex

(Vectastain® ABC Kit, Vector Laboratories, Burlingame, USA) and those for CD4 by

the SignalStain® Boost IHC Detection Reagent (New England Biolabs, Frankfurt,

Germany). Diaminobenzidine was used as chromogen and intensified by a short

incubation with 0.01 % osmic acid. Sections were counterstained with 2 % methylene

green except those for Ki-67 and factor VIII that were counterstained with Mayer´s

hematoxylin and cover slipped with Kaiser’s glycerin gelatin. Sections containing the

specific cell types were included as positive controls and sections where the first

antibody was replaced by an antibody against unrelated antigen as negative controls.

5.3.4 Image analysis of the size and cellular subsets in granulomas

Measurement of granuloma areas in sections was used as estimate of granuloma

size. This was performed with Image J for all granulomas of all stages in selected

JPP, IPP and ILN. Granulomas were measured by manual tracing of their outline in

HE-stained sections. Stages 1 and 2 were measured at x 100 magnification, stage 3

on x 1 or x 50 magnification depending on size, stages 4 and 5 x 1 magnification and

stage 6 on x 1 or x 50 magnification depending on size. Magnification of factor 1 was

achieved by scanning whole slides with a flatbed scanner (Epson V700, Seiko Epson

Corporation, Suwa, Japan). Numbers of CD4+, CD8+, CD25+, TcR1-N24+, CD79α+,

CD68+, Ki-67+ cells, endothelial cells and neutrophils were semi-quantitatively

assessed at x 400 magnification in a reference area (RA) of 2359 µm2 measured by

an ocular grid. Cell numbers were converted to scores (Tab. 5.3). MGCs were

differentiated as small MGCs (<10 nuclei) and large MGCs (>10 nuclei).

Representative images were captured using a Leica DMR microscope (Leica,

Page 95: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

91

Germany) equipped with an Olympus DP71 video camera (Olympus, Germany) and

imaging software (Cell F, Version 2.6, Olympus, Germany).

Table 5.2: Antisera used for immunohistochemistry

target/ cells

epitope/ antigen

clone dilution supplier secondary antibody

method

T helper cells

CD4 GC1A 1:50 VMRD, Pullman, USA, distrib.

Labor Diagnostik Leipzig, Germany

Boost Kita indirect immuno-

peroxidase (IP)

cytotoxic T cells

CD8 CC63 un-diluted

European Cell Culture Collection,

Salisbury, UK

sheep anti-mouse IgGb

indirect IP

γδ T cells TcR1-N24 (δ chain)

GB21A 1:400 VMRD sheep anti-mouse IgGb

indirect IP

activated T cells

CD25 (IL-2Rα)

CACT 116A

1:500 VMRD biotin. goat anti-mouse IgGc, ABCd

avidin-biotin

complex B cells, plasma

cells

CD79α HM57 1:200 AbD Serotec, Düsseldorf, Germany

biotin. goat anti-mouse IgGc, ABCd

avidin-biotin

complex macro-phages

CD68 EBM11 1:50 Dako, Glostrup, Denmark

sheep anti-mouse IgGb

indirect IP

MHC-II MHC-II H42A 1:4000 European Cell Culture Collection,

Salisbury, UK

sheep anti-mouse IgGb

indirect IP

proliferat-ing cells

Ki-67 MIB-1 1:200 Dako, Glostrup, Denmark

sheep anti-mouse IgGb

indirect IP

endothel-ial cells

Van Willebrand

Factor

--- 1:800, 1:2000*

Dako, Glostrup, Denmark

goat anti-rabbit IgGe

indirect IP

a, SignalStain® Boost, New England Biolabs GmbH, Frankfurt, Germany; b,

NA931V, GE Healthcare Europe GmbH, Freiburg, c, GE Healthcare Europe GmbH,

Freiburg, Germany; d, Vector Laboratories, Inc., Burlingame, USA; distributed by

Biologo, Konshagen, Germany, e, Jackson Immunoresearch; distributed by Dianova,

Hamburg, Germany; * dilution of 1:800 used in oGALT and 1:2000 used in ILN

Page 96: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

92

Table 5.3 Semi-quantitative assessment of different immunohistochemically labeled

cell types, neutrophils and AFB

score - (+) + ++ +++ ++++

none scattered single few many numerous

number of lymphocytes, plasma cells, proliferating

cells,neutrophils/RA

0 1-2 3-8 9-14 15-20 >20

number of macrophages/RA

0 1-2 3-6 7-10 11-15 >15

number of AFB 0 1-3 4-15 16-50 50-200 >200

RA, reference area (2359 µm²)

5.4. Results

In the following, first the cellular composition of granulomas of stages 1-6 will be

described and then the distribution at 2-3 mpi versus 13 mpi in oGALT versus ILN.

5.4.1 Cellular composition of granulomas

5.4.1.1 Stage 1 granulomas (initial, Fig. 5.1 A-F)

They were composed of small clusters of CD68+ epitheloid macrophages (Fig. 5.1A,

B) which were weakly MHC-II+ (Fig. 5.1C). Few to single CD4+ T lymphocytes (Fig.

5.1D), scattered CD8+ T lymphocytes, γδ T lymphocytes, neutrophils and proliferating

cells, but no B lymphocytes (Fig. 5.1E) were present between the epitheloid

macrophages. Single strongly MHC-II+ cells with dendritic morphology (Fig. 5.1C)

surrounded the granulomas. Stage 1 granulomas were often observed in the

paracortex and thus surrounded by many to numerous CD4+ (Fig. 5.1D) and few to

many CD8+ T lymphocytes. If present, scattered AFB were detected in the cytoplasm

of epitheloid macrophages (Fig. 5.1F).

5.4.1.2 Stage 2 granuloma (solid, minimal necrosis)

Stage 2 granulomas were slightly larger (Tab. 5.4), but similar in cellular composition

to stage 1 granulomas (Fig. 5.2 A-E). Single small MGCs were occasionally seen at

13 mpi. A few areas of single to few necrotic cells (Fig 5.2A), predominantly CD68+

Page 97: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

93

epitheloid macrophages (Fig. 5.2B), were present. Thin collagen fibers occurred

throughout the granuloma in a pattern similar in the surrounding tissue. If present,

scattered AFB were detected in the cytoplasm of epitheloid macrophages (Fig. 5.2F).

5.4.1.3 Stage 3 (monocentric necrosis and mineralization)

They were larger than stage 1 and 2 granulomas (Tab. 5.4), often round structures,

composed of a central area with caseous necrosis and mineralization surrounded by

distinct layers of inflammatory cells (Fig. 5.3A). Few thin collagen fibers were radially

orientated in the inner granulomatous zone (Fig. 5.3H).

No distinct cell types could be discerned in the area of central caseous necrosis and

mineralization, but if present scattered AFB were found. At the edge of necrosis,

epitheloid macrophages and MGC were degenerated and scattered AFB were

detected in the cytoplasm of a few of these epitheloid macrophages. The necrosis

was completely surrounded by a wide zone of granulomatous infiltrate composed of

many CD68+ epitheloid macrophages (Fig. 5.3B), few CD4+ T lymphocytes (Fig.

5.3D), single CD8+ T lymphocytes, scattered γδ T lymphocytes, CD25+ cells , B

lymphocytes and plasma cells (Fig. 5.3E), and single neutrophils. A small number of

large and moderate numbers of small MGC´s were observed (Fig. 5.3B). A few

intensely MHC-II+ cells with dendritic morphology (Fig. 5.3C) were observed between

the epitheloid macrophages and lymphocytes. Very few cells of the infiltrate were

Ki67+ (Fig. 5.3F). Cells were embedded in a network of endothelial cells, capillaries

and single venules (Fig. 5.3G).

The granulomatous infiltrate was surrounded by an incomplete variable thick zone of

lymphocytes resembling tertiary lymphoid tissue (3rd LT) in an early developmental

phase. The 3rd LT was organized into lymphoid follicle-like structures with numerous

B lymphocytes, single plasma cells (Fig. 5.3E), CD4+ T lymphocytes (Fig. 5.3D),

CD68+ macrophages (Fig. 5.3B), scattered CD8+ T lymphocytes and occasional foci

with increased proliferation, and interfollicular areas (IFA) with many CD4+ T

lymphocytes (Fig. 5.3D), few CD8+ T lymphocytes, single γδ T lymphocytes, and

scattered intensely MHC-II+ dendritic cells (Fig. 5.3C), CD25+ cells and CD68+

macrophages. There were few capillaries, venules, arterioles (Fig. 5.3G) and a few

Page 98: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

94

vessels with cuboidal endothelium possibly representing high endothelial venules.

Scattered AFB were associated with some macrophages in the B-cell rich zone of the

3rd LT in one granuloma (Fig. 5.3I).

Multiple, rather small granulomas with almost completely mineralized centers

frequently lacking a zone of granulomatous infiltrate, but surrounded by a few layers

of collagen fibers and occasionally associated with 3rd LT were observed

predominantly in ILN and scarcely in oGALT (Tab. 5.4). Although, they were

morphologically distinct, they were included under stage 3 granulomas.

5.4.1.4 Stage 4 (multicentric necrosis and mineralization)

Stage 4 granulomas were highly variable in size, overall much larger than stage 3

granulomas and irregular in shape (Tab. 5.4). They were characterized by extensive

multicentric caseous necrosis and mineralization surrounded by distinct layers of

inflammatory cells and collagen fibers (Fig. 5.4A).

The zone of granulomatous infiltrate surrounding the necrosis was variably thick, with

scattered large and few small MGCs. The cellular composition was similar as in stage

3 granulomas (Fig. 5.4B, D, E), but the amount of neutrophils, endothelial cells and

capillaries was lower. Scattered AFB were seen in the areas of necrosis and

mineralization and in the cytoplasm of single epitheloid macrophages of the

granulomatous infiltrates as described for granulomas of stage 3.

The zone of granulomatous infiltrate was partly or completely enclosed by an

irregular zone of connective tissue with mature collagen fibers (Fig. 5.4C) which was

especially thick in the proximity of pre-existing connective tissue, e.g. the capsule of

lymph nodes. Many intensively labeled MHC-II+ cells, single CD4+ T lymphocytes (Fig.

5.4D), CD68+ macrophages (Fig. 5.4B) and scattered CD8+ T cells, γδ T

lymphocytes, CD25+ cells, B lymphocytes and plasma cells (Fig. 5.4E) and

proliferative cells (Fig. 5.4F) were present between the strands of connective tissue.

Cell numbers were slightly increased immediately adjacent to the granulomatous

infiltrate. In addition, single capillaries and few endothelial cells could be

differentiated.

Page 99: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

95

Well differentiated 3rd LT was observed at the periphery of the connective tissue (Fig.

5.4D-F). It consisted of lymphoid follicles with proliferating B cells (Fig. 5.4E, F)

forming light and dark zones; thus indicating the presence of germinal centers (GC).

GC of 3rd LT were oriented with the light zone towards the center of granulomas and

thus distinct from GC in the peripheral cortex that were directed towards the

subcapsular sinus. Lymphoid follicles were separated by large IFA with a cellular

composition as described for granulomas of stage 3 (Fig. 5.4D), but increased

numbers of high endothelial venules and capillaries.

5.4.1.5 Stage 5 (fibrotic organization)

Stage 5 granulomas were comparable in size to stage 4 granulomas (Tab. 5.4).

Small multicentric islands of necrosis and mineralization were surrounded by

extensive connective tissue (Fig. 5.5A, C) and 3rd LT, whereas the granulomatous

infiltrate was minimal (Fig. 5.5A) or missing.

When present, the small zones of granulomatous infiltrate consisted of many CD68+

epitheloid macrophages, few CD4+ T lymphocytes and γδ T lymphocytes (Fig. 5.5B)

as well as scattered small MGCs, CD8+ T lymphocytes, CD25+ cells, B lymphocytes,

plasma cells and neutrophils. Scattered AFB were seen in the islands of necrosis and

mineralization and in the cytoplasm of single epitheloid macrophages in the zone of

granulomatous infiltrate (Fig. 5.5F). In the extensive zone of connective tissue, a

similar distribution and number of inflammatory cells as described in stage 4

granulomas were seen. The amount of endothelial cells and capillaries was very low

in these zones (Fig. 5.5E). The morphology of 3rd LT was comparable to that in stage

4 granulomas (Fig. 5.5D).

5.4.1.6 Stage 6 (extensive necrosis and minor mineralization)

These granulomas varied markedly in size (Tab. 5.4). They were characterized by

extensive zones of caseous necrosis with multiple small foci of mineralization which

were surrounded by an irregular, variably thick zone of inflammatory cells not well

demarcated from the pre-existing lymphoid tissue (Fig. 5.6A).

Page 100: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

96

The necrosis was not uniform, but consisted of areas with nuclear fragments and

ghost cells, and areas interspersed with numerous confluent vacuoles of variable

size (Fig. 5.6B). Small clusters of few to many extracellular AFB were frequently seen

in the necrotic material (Fig. 5.6F). At the edge of the necrosis, there was a zone of

degenerated cells, predominantly CD68+ epitheloid macrophages (Fig. 5.6D),

intermixed with single CD4+ T lymphocytes (Fig. 5.6E), CD8+, γδ T lymphocytes,

CD25+ cells, B cells, plasma cells and neutrophils. Multifocally, intense MHC-II+

dendrites (Fig 5.6C) were interspersed between these cells. Few AFB were found

extracellularly between cellular fragments (Fig. 5.6G) and scattered AFB were

present in epitheloid macrophages. Scattered Ki-67+ cells and single endothelial cells

could be found.

The necrosis was surrounded by a granulomatous infiltrate dominated of many CD4+

T lymphocytes (Fig. 5.6E), small clusters of CD68+ epitheloid macrophages (Fig.

5.6D) and remnants of pre-existing lymphoid tissue. There were few B lymphocytes,

scattered plasma cells, single CD8+ T lymphocytes and scattered γδ T lymphocytes,

CD25+ cells and neutrophils. Clusters of epitheloid macrophages were surrounded by

intensely MHC-II+ dendritic cells. Single endothelial cells, capillaries and venules

were present. Haphazardly oriented, immature collagen fibers were present from the

edge of the necrosis throughout the granulomatous infiltrate.

5.4.2 Distribution of granuloma stages 1-6 in oGALT versus ILN of goats at 2-3

mpi versus 13 mpi

Many stage 1 and 2 granulomas were seen in the cortex and to a lesser extent in

medullary regions in ILN at 2-3 mpi, whereas only few were found in oGALT and ILN

at 13 mpi (Tab. 5.4). Stage 3 granulomas were larger in submucosal areas of IPP

than in ILN (Tab. 5.4). Large stage 4 and 5 granulomas were only seen in ILN at 13

mpi (Tab. 5.4). Stage 6 granulomas of varying size were uniquely found in ILN at 2-3

mpi (Tab. 5.4). Figure 5.7 proposes a scheme of granuloma development during

infection of goats with MAH.

Page 101: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

97

5.4.3 Ulcerative and granulomatous lesions in oGALT at 2-3 mpi

In the goats necropsied at 2-3 mpi, granulomatous infiltrates were seen in oGALT,

especially the IPP. There were similarities to stage 6 granulomas, but no distinct

granuloma formation as described above. This is most likely due to the extensive

ulceration of the overlying mucosa. Therefore this lesion is described separately. An

extensive, T cell-dominated infiltrate had replaced large parts of the preexisting

oGALT and the mucosa was extensively ulcerated. The infiltrate was characterized

by many CD4+, few CD8+, single γδ T lymphocytes, B lymphocytes and plasma cells

as well as scattered CD25+ cells and variable numbers of neutrophils. Clusters of

single to few CD68+ epitheloid macrophages were present in the infiltrate. They were

surrounded by intensely MHC-II+ cells with dendritic morphology. Scattered AFB

could be seen in single epitheloid macrophages.

Page 102: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

98

Table 5.4 Number and areas of different granuloma stages in goats infected with

MAH at 2-3 mpi and 13 mpi in oGALT and ILN.

GS months

post inocul.

location number (n)

associated with 3rd LT

minimal area (mm2)

maximal area (mm2)

median area (mm2)

1 2-3

oGALT 0 0/0 - - - ILN 87 0/87 0.003 0.05 0.021

13 oGALT 1 0/1 0.034 0.034 0.034

ILN 6 0/6 0.096 0.23 0.124

2 2-3

oGALT 0 0/0 - - - ILN 13 0/13 0.097 0.46 0.194

13 oGALT 2 0/2 0.095 1.07 0.5825

ILN 4 0/4 0.118 0.216 0.1425

3 2-3

oGALT 0 0/0 - - - ILN 0 0/0 - - -

13 oGALT 22 13/22 0.064 3.467 0.9245

ILN 32 26/32 0.109 1.81 0.2775

4 2-3

oGALT 0 0/0 - - - ILN 0 0/0 - - -

13 oGALT 0 0/0 - - -

ILN 10 10/10 0.215 43.79 6.331

5 2-3

oGALT 0 0/0 - - - ILN 0 0/0 - - -

13 oGALT 0 0/0 - - -

ILN 5 5/5 2.671 58.278 5.159

6 2-3

oGALT 0 0/0 - - - ILN 28 0/28 0.357 34.03 0.8985

13 oGALT 0 0/0 - - -

ILN 0 0/0 - - -

GS, granuloma stage; inocul., inoculation; oGALT, organized gut-associated

lymphoid tissue; ILN, intestinal lymph nodes

Page 103: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

99

Page 104: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

100

Figure 5.1

Consecutive sections of stage 1 granulomas (indicated as hatched lines) in the

interfollicular areas of the cortex in ILN. (A) Groups of epitheloid macrophages

surrounded by lymphocytes in the paracortex. (B) Stage 1 granulomas consist

predominantly of CD68+ epitheloid macrophages with abundant cytoplasm. Slender

CD68+ macrophages are present in the surrounding paracortex. (C) Epitheloid

macrophages are weakly MHC-II+ and scattered intensely MHC-II+ cells with dendritic

morphology are seen in the periphery (arrow). (D) The paracortex is dominated by

CD4+ T lymphocytes, single CD4+ T lymphocytes are inside the granuloma obscuring

its delineation. (E) Clusters of many B lymphocytes and scattered plasma cells are

seen around the granuloma but not within. (F) One AFB (arrow) in an epitheloid

macrophage.

Page 105: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

101

Hematoxylin and eosin staining in panel A, IHC in B-E, Kinyoun stain in panel F,

scale bar = 100 µm in panels A-E, scale bar = 20 µm in panel F.

Page 106: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

102

Figure 5.2

Consecutive sections of stage 2 granuloma at the transition between paracortex and

medulla in an ILN. (A) Stage 2 granuloma consists predominantly of epitheloid

macrophages and small areas of necrotic cells (arrow). (B) Epitheloid macrophages

in the granuloma have abundant CD68+ cytoplasm; some of them are necrotic

(arrow). (C) Epitheloid macrophages are weakly MHC-II+, intensively MHC-II+ cells

are present at the edge and throughout the granuloma some with dendritic

morphology (arrow). (D) There are single CD4+ T lymphocytes in the granuloma. (E)

Single γδ T lymphocytes are seen in the paracortex around the granuloma but not

within. (F) Extracellular AFB (arrow) next to cellular fragments.

Page 107: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

103

Hematoxylin and eosin staining in panel A, IHC B-E; Kinyoun stain in panel F, scale

bar = 200 µm in panels A-E, scale bar = 20 µm in panel F.

Page 108: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

104

Figure 5.3

Consecutive sections of stage 3 granuloma in an IPP with central necrosis and

mineralization (I), a layer of granulomatous infiltrate (II) and tertiary lymphoid tissue

(3rd LT, III). (A) Well-structured granuloma with the 3 distinct layers (I-III) as

mentioned above in the submucosa (SM). The layer of granulomatous infiltrate

extents focally (*) through the 3rd LT. (B) Epitheloid macrophages with abundant

CD68+ cytoplasm in II, a MGC is indicated (arrow). (C) Epitheloid macrophages and

MGCs cells are weakly MHC-II+, intensely MHC-II+ cells with dendritic morphology

are multifocally present in II (*) and scattered in the periphery of III. (D) Few CD4+ T

lymphocytes are present in II. There is a distinct distribution of CD4+ T lymphocytes

in III forming interfollicular areas (*) and lymphoid follicle-like structures (LF). (E)

Distinct distribution of B lymphocytes in III forming lymphoid follicle-like structures.

Scattered B lymphocytes and plasma cells in II and in the periphery of III. (F) Single

Ki-67+ proliferative cells in II and III, no increase in lymphoid follicle-like structures

(LF). (G) Granulomatous layer is highly vascularized. There are multifocal arterioles

and venules in III and many in the submucosa (SM) next to the granuloma. (H) Few

weakly stained collagen fibers (light blue) in II. Granuloma is surrounded by mature

collagen of the submucosa (SM). (I) Scattered AFB (arrow, exampl.) in a

macrophage in a lymphoid follicle-like structure of 3rd LT.

Page 109: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

105

Hematoxylin and eosin staining in panel A; IHC in panel B-G, Azan stain in panel H;

Kinyoun stain in panel I, scale bar = 200 µm in panels A-H, scale bar = 20 µm in

panel I.

Page 110: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

106

Figure 5.4

Consecutive section of a stage 4 granuloma in an ILN with central necrosis and

mineralization (I), with focal granulomatous infiltrate (II), variably thick layer of

connective tissue (III) and tertiary lymphoid tissue (3rd LT, IV), delineated by hatched

lines. (A) There are only fragments of zone I. The loss of necrotic tissue is a

preparation artifact caused by extensive mineralization. A focal area of

granulomatous infiltrate (II) is surrounded by an irregularly thick layer of connective

tissue (III). 3rd LT (IV) is difficult to distinguish from preexisting lymphoid tissue in the

HE stain. (B) Many CD68+ epitheloid macrophages with abundant cytoplasm are

present in II and single slender CD68+ macrophages (arrow) in III. (C) The layer of

connective tissue (III) consists of mature collagen fibers; it separates the 3rd LT (IV)

from the granuloma center. (D) Few CD4+ T lymphocytes are seen in II and single in

III. Compartments of 3rd LT are more distinct than in stage 3 granulomas with large

CD4+ T lymphocyte-rich interfollicular areas (*) and lymphoid follicles (LF) with single

CD4+ T lymphocytes. (E) Lymphoid follicles (LF) of 3rd LT (IV) are rich in B

lymphocytes and germinal centers can be distinguished. All LF of the 3rd LT are

oriented towards the granuloma center indicating the direction of antigenic influx.

Scattered B lymphocytes and plasma cells (arrow) are seen in II and III. (F) Ki-67+

labeling shows the presence of well-organized germinal centers in lymphoid follicles

(LF) of 3rd LT (IV) with light zone (a) and dark zone (b). Single and scattered Ki-67+

cells are found in II and III, respectively.

Page 111: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

107

Hematoxylin and eosin staining in panel A; IHC in panel B, D-F, Azan stain in panel

C, scale bar = 500 µm in panel A, scale bar = 200 µm in panels B-F.

Page 112: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

108

Figure 5.5

Consecutive sections of a stage 5 granuloma in an ILN with circumscribed necrosis

and mineralization (I), minimal granulomatous infiltrate (II), surrounded by extensive

connective tissue (III) and surrounded by tertiary lymphoid tissue (3rd LT, IV). (A)

Stage 5 granuloma consists predominantly of connective tissue (III). (B) Increased γδ

T lymphocytes are present in II. (C) Connective tissue is formed by mature (dark

blue) collagen fibers. (D) Lymphoid follicle (LF) of the 3rd LT (IV) is oriented with the

light zone (a) towards the granuloma center. (E) Vascularization is minimal in the

granulomatous infiltrate (II) and connective tissue (III) as indicated by scattered

endothelial cells (arrow, exampl.) and capillaries (arrowhead, exampl.), respectively.

There are a few venules and arterioles in the 3rd LT (IV). (F) An AFB is present in an

epitheloid macrophage (arrow) in II.

Page 113: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

109

Hematoxylin and eosin staining in panel A; IHC in panels B, D, E; Azan stain in

panel C, Kinyoun stain in panel F, scale bar = 500 µm in panel A, scale bar = 200 µm

in panels B-E, scale bar = 20 µm in panel F.

Page 114: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

110

Figure 5.6

Consecutive sections of a stage 6 granuloma in an ILN with extensive caseous

necrosis and mineralization (I), a transition zone of degenerated inflammatory cells

(II) and a zone of granulomatous infiltrate (III). (A) Extensive caseous necrosis with

multiple areas of mineralization (*, exampl., I). A delicate irregularly thick transition

zone of degenerated inflammatory cells (in II), surrounded by a lymphocyte-rich

granulomatous infiltrate (III). (B) Higher magnification of necrosis as indicated by the

black rectangle in A: dense, granular, eosinophilic material with nuclear fragments

(a), small focal mineralization (b) and eosinophilic material interspersed with

numerous often confluent vacuoles (c). (C) Many intensely MHC-II+ dendrites

(arrows, exampl.) are present between inflammatory cells in the transition zone (II).

(D) Single (arrow, exampl.) and multiple clusters of CD68+ epitheloid macrophages

with abundant cytoplasm in II and III, respectively. (E) Single and numerous CD4+ T

lymphocytes in II and III, respectively. (F) Many occasionally clustered extracellular

AFB inside the vacuolated center of the necrosis (I). (G) Few extracellular AFB (↙)

between cellular debris in the transition zone (II).

Page 115: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

111

Hematoxylin and eosin staining in panel A and B, IHC in panels C-E, Kinyoun stain

in panel F and G, scale bar = 500 µm in panel A; scale bar = 200 µm in panels B, D-

E; scale bar = 100 µm in panel C, scale bar = 20 µm in panels F and G.

Page 116: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

112

Figure 5.7 Scheme of granuloma development during infection of goats with MAH

Page 117: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

113

5.5 Discussion

Granulomas with a wide variety of morphological characteristics were observed in

goats after oral inoculation with MAH (Schinköthe et. al, submitted). In the current

study, granulomas were classified based on extent of necrosis, mineralization,

inflammatory cell infiltrate, vascularization and fibrosis as well as overall organization

and cellular composition of the inflammatory infiltrate. Classification of granulomas is

important, because it generates information about the pathogenesis of infection and

the host response at a local level. Stages of granulomas can frequently be found in

an infected individual, even within the same organ, and may reflect differences in

microenvironment or age of the lesion (PALMER et al. 2007; LENAERTS et al.

2015).

Staging has been used in experimental mouse models (RHOADES et al. 1997),

guinea pig models (TURNER et al. 2003) and more extensively in natural and

experimental bovine TB (WANGOO et al. 2005; PALMER et al. 2007; ARANDAY-

CORTES et al. 2013; MENIN et al. 2013). It is the basis for the comparison of host-

pathogen interactions in different species and different mycobacterial strains

(WANGOO et al. 2005) and for testing the protective effect of vaccine candidates

(LIEBANA et al. 2007; MENIN et al. 2013). Unfortunately, assessment of granuloma

stages varies between studies most likely due to differences in species examined,

experimental design and histologic and immunologic methods applied. The findings

in the goats infected with MAH were most closely reflected in the staging used for

cattle infected with Mycobacterium bovis by Wangoo et al. (WANGOO et al. 2005).

The additional granuloma stages seen in this investigation were added to the end of

the scheme by Wangoo et al. as the granulomas with extensive necrosis and limited

organization (stage 6 granulomas) and the granulomas with fibrosis as main

characteristic (stage 5 granulomas)

In the following the characteristics of the granulomas seen in goats infected with

MAH will be highlighted and compared with the findings especially in granulomas

seen in cattle and humans (CASSIDY et al. 2001; ULRICHS et al. 2004; ULRICHS et

al. 2005; WANGOO et al. 2005; ULRICHS u. KAUFMANN 2006; PALMER et al.

Page 118: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

114

2007; ARANDAY-CORTES et al. 2013). The functional implications of the

morphological findings will be discussed.

Stage 1 granulomas formed predominantly by epitheloid macrophages and

containing very low numbers of AFB were seen as initial stages of infection under our

experimental conditions in goats predominantly in ILN at 2-3 mpi, but also at 13 mpi.

This indicates that continuous dissemination occurs that may originate from lesions in

oGALT or ILN. Comparable lesions have been recognized usually after 3 mpi in

experimental bovine TB studies (WANGOO et al. 2005; PALMER et al. 2007;

ARANDAY-CORTES et al. 2013). In the first two month post inoculation, studies in

guinea pigs and cattle reported pseudo-eosinophilic and degenerated neutrophils,

macrophages and MGCs as additional morphological features and many intra- and

extracellular AFB (CASSIDY et al. 2001; TURNER et al. 2003; PALMER et al. 2007).

A consecutive study in cattle was able to demonstrate moderate to numerous AFB in

stage 1 granulomas in bovine lung and lymph nodes between 15-60 dpi, while few to

no AFB were seen from 90 dpi on (PALMER et al. 2007). The latter type of

granulomas were named satellite granulomas (PALMER et al. 2007). They resemble

most closely the type 1 granulomas seen in the goats infected with MAH. The

difference in the number of AFB is most likely due to the increased activation of

macrophages after the onset of adaptive immunity (PALMER et al. 2007). As

reported in bovine TB (PALMER et al. 2007), few T lymphocytes, including CD4+,

CD8+, γδ and CD25+ cells in decreasing number, were present between the

epitheloid macrophages. Since stage 1 granulomas were frequently present in the

paracortex, it remains unresolved if these cells represent pre-existing lymphoid tissue

or a reactive infiltrate. A prominent boundary zone with abundant CD8+ T

lymphocytes and CD25+ cells as described in bovine TB (LIEBANA et al. 2007) was

not observed.

The cellular composition of stage 2 granulomas was highly similar to that of stage 1

granulomas, but in addition small areas of necrosis were seen. The extent of

necrosis was very limited in our material compared to that described in respiratory

infection of cattle with M. bovis (WANGOO et al. 2005; ARANDAY-CORTES et al.

2013). The rare AFB detected were associated with the areas of necrosis and cellular

Page 119: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

115

debris. These foci of necrosis are most likely the starting points of the more extensive

necrosis found in stage 3, 4, 5 and 6 granulomas. The infiltrate of dendritic cells

indicates antigen presentation and initiation of an adaptive immune response. There

was no fibrous capsule as reported in some other studies (PALMER et al. 2007;

ARANDAY-CORTES et al. 2013).

There were several granuloma stages (3, 4 and 6) with more extensive necrosis; it

remains unclear, if there is a progression of stage 6 granulomas to stage 3 or 4

granulomas or if stage 6 granulomas represent a “dead end” development due to a

dysbalanced immune response. Stage 6 granulomas were only seen in lymph nodes

from severely sick goats at 2-3 mpi. Comparable lesions were not reported in other

studies, but they have some features of transmissive granulomas described as

consequence of a Th1/Th2 dysbalance (EHLERS u. SCHAIBLE 2012). Stage 6

granulomas were characterized by extensive caseous necrosis of variable

appearance: homogeneous eosinophilic material, nuclear remnants and multiple

confluent vacuoles. Vacuoles were also reported in a mouse study and were

associated with cholesterol byproducts that accumulated inside the necrosis

(RHOADES et al. 1997). Mycobacteria can shift their metabolism towards processing

cholesterol under nutrient stress (EHLERS u. SCHAIBLE 2012) as might have

happened, since many AFB were present in the necrosis. The necrosis was

surrounded by a wide zone of cellular debris and degenerate inflammatory cells

indicating rapid expansion of the lesion. The granulomatous infiltrate that followed

consisted predominantly of CD4+ T lymphocytes and small groups of epitheloid

macrophages. Experimental infection with M. avium species in mice demonstrated

that a protective immune response is primarily driven by CD4+ T lymphocytes, while

CD8+ and γδ T lymphocytes did not significantly influence the host pathogen

interaction (APPELBERG et al. 1994; PETROFSKY u. BERMUDEZ 2005). In the

case of goats infected with MAH, the large number of CD4+ T lymphocytes can be

interpreted as excessive immune response. Besides other cytokines, CD4+ T

lymphocytes may secrete IFN-γ, which activates macrophages to secrete TNF-α

(DORHOI u. KAUFMANN 2014). Excessive IFN-γ and TNF-α secretion results in a

hyper-inflammatory milieu that leads to tissue damage. This was seen as wide zone

Page 120: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

116

of cell degeneration. Mycobacteria released from degenerated cells may infect

recruited macrophages thus establishing new foci of infection and maintaining

granuloma growth similar to that described in the zebrafish model (RAMAKRISHNAN

2012). The overwhelming pro-inflammatory immune response might also be the

cause why structured tuberculous granulomas did not develop and extensive fibrosis

was missing. This would have required a balanced immune response as seen in

latent tuberculosis (FLYNN et al. 2011).

In goats with stage 6 granulomas, comparable lesions were seen in oGALT. The

excessive immune response with abundant numbers of CD4+ T lymphocytes had

most likely caused extensive loss of mucosa including necrotic tissue. Thus there

was little necrosis, but predominantly an infiltrate of CD4+ T lymphocytes and small

groups of epitheloid macrophages as described in the periphery of stage 6

granulomas. The loss of mucosal barrier resulted in an influx of neutrophils. These

lesions in oGALT were not included in the granuloma stages, because they were

rather diffuse. There is, however, some resemblance to cavitating lesions described

in the lung (BASARABA 2008).

Stage 3, 4 and 5 granulomas are highly organized structures observed only in

clinically healthy goats at 13 mpi. Others reported lesions similar to stage 3

granulomas in experimental bovine TB as early as 42 dpi (CASSIDY et al. 2001).

Stage 3 granulomas were seen most frequently, both in oGALT and ILN. Since there

was a central necrosis, the surrounding zones of predominantly epitheloid

macrophages and of lymphocytes were easy to recognize. Comparable granulomas

are also frequent in late stages of human pulmonary TB and in bovine and guinea pig

TB (TURNER et al. 2003; ULRICHS et al. 2004; WANGOO et al. 2005; LIEBANA et

al. 2007; PALMER et al. 2007; ARANDAY-CORTES et al. 2013).

A distinct feature of stage 3, 4, and 5 granulomas was the zone of lymphocytes in the

periphery that became progressively organized. Follicle-like B lymphocyte clusters

and T lymphocyte-rich areas were seen in stage 3 granulomas. Follicles with

germinal centers organized as light and dark zones and distinct interfollicular areas

with descending numbers of CD4+, CD8+, γδ T lymphocytes and dendritic cells were

present in stage 4 and 5 granulomas. This compartmentalization fulfills the definition

Page 121: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

117

of ectopic lymphoid tissue that develops in association with chronic infectious

diseases and cancer (ULRICHS et al. 2004; PITZALIS et al. 2014). Ectopic lymphoid

tissue also named 3rd LT is a hallmark of human pulmonary TB granulomas

(ULRICHS et al. 2004; PITZALIS et al. 2014). Aggregates of B lymphocytes and

compartmentalization of different leukocyte subtypes were reported in cattle and

goats with experimental and natural mycobacterial infections (WANGOO et al. 2005;

SANCHEZ et al. 2011; ARANDAY-CORTES et al. 2013), but rarely interpreted as 3rd

LT. However, the potential control of mycobacterial infection by a local and

coordinated immune response was discussed (ARANDAY-CORTES et al. 2013). A

differentiation from pre-existing lymphoid tissue in lymph nodes is possible by the

distinct orientation of the 3rd LT toward the granuloma center.

It has been suggested that 3rd LT is essential for the containment of mycobacteria

within the granuloma (ULRICHS et al. 2004; ULRICHS u. KAUFMANN 2006). In this

context the relevance of cytolytic killing of infected macrophages by CD8+ T

lymphocytes and their role for the development of necrosis was questioned, since

they were primarily found in the interfollicular areas of the 3rd LT in human pulmonary

TB granulomas and not close to the necrosis (ULRICHS et al. 2004; ULRICHS u.

KAUFMANN 2006). This is further confirmed by the findings in the goats infected with

MAH and in guinea pigs with experimental TB (TURNER et al. 2003). As reported for

3rd LT in human pulmonary TB, macrophages in the lymphoid follicles of the goats

contained mycobacteria (ULRICHS et al. 2004; ULRICHS u. KAUFMANN 2006). The

presence of mycobacteria in the lymphoid follicles may indicate a direct initiation of B

lymphocyte responses. Recent reports suggested that B lymphocytes can present

peptide antigens via MHC-II molecules, prime T lymphocytes to secret a variety of

cytokines and chemokines, and thus modulate the granulomatous inflammation,

cytokine production and influence the control of TB infection (FLYNN et al. 2011;

CHAN et al. 2014; HOFF et al. 2015). On the other hand, the dendritic cells seen

around the necrosis, in the layer of granulomatous infiltrate and in the interfollicular

areas of 3rd LT of the goats infected with MAH, might have migrated from the center

of the granuloma to the 3rd LT to activate T lymphocytes. The minimal numbers of

Page 122: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

118

intra- and extracellular AFB in granulomas with 3rd LT indicate that the immune

response was effective in limiting proliferation of mycobacteria.

A zone of connective tissue between the granulomatous infiltrate and the 3rd LT was

a distinct characteristic of stage 4 and 5 granulomas, but not seen in stage 3

granulomas of the goats infected with MAH. This agrees with observations in human

TB (ULRICHS et al. 2004), but not in bovine TB where complete fibrous capsules are

already seen in stage 3 granulomas (WANGOO et al. 2005; ARANDAY-CORTES et

al. 2013). In stage 4 granulomas, there was extensive multicentric necrosis and

moderate fibrosis which most likely progressed to the small circumscribed, heavily

mineralized necrosis and extensive fibrosis of stage 5 granulomas. The progress of

mineralization and fibrosis has been considered as favorable for the host and as

healing process (BASARABA 2008).

To gain knowledge about the origin of the inflammatory cells the vascularization and

the cellular proliferation in the different compartments of the granulomas were

investigated. Only few proliferating cells were seen in the granulomas except for 3rd

LT indicating limited local expansion of inflammatory cell populations. Differences

were seen in the vascularization between granuloma stages due to differences in the

vascularization of the different zones. The zone of granulomatous infiltrate was highly

vascularized, the zone of connective tissue almost devoid of blood vessels and the

3rd LT vascularized similar to regular lymphoid tissue. Since the layer of

granulomatous infiltrate was most extensive in stage 3 granulomas, they were better

vascularized than stage 4 and 5 granulomas. Stage 3 granulomas resembled in this

respect tuberculomas in human pulmonary TB (ULRICHS et al. 2005). The authors

concluded that vascularization promotes efficient cross talk between host and M.

tuberculosis due to the supply with oxygen and nutrients which renders highly

vascularized granulomas as non-progressive TB lesions. On the other hand, the high

vascularization of type 3 granulomas may allow a continuous influx of inflammatory

cells including macrophages that may become infected by mycobacteria.

Page 123: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

119

5.6 Conclusion

After oral inoculation of goats with MAH, a wide variety of granulomas developed.

The lesions observed in the severely sick goats at 2-3 mpi are representative of an

exuberant immune response causing marked tissue damage. The lesions observed

in the surviving goats at 13 mpi reflect the heterogeneity of lesions seen in goats and

cattle with MTC TB, and humans with pulmonary TB (VAN RHIJN et al. 2008;

LENAERTS et al. 2015). Especially the regularly observed 3rd LT may be an

important morphological characteristic to assess the control of mycobacterial

infections by the host (ULRICHS et al. 2004; ULRICHS et al. 2005; ARANDAY-

CORTES et al. 2013). Thus, the experimental infection of goats with MAH has certain

advantages: (1) in comparison to cattle, goats are much smaller in size facilitating

housing, feeding and handling (DE VAL PEREZ et al. 2011; GONZALEZ-

JUARRERO et al. 2013), (2) infection with MAH as BSL 2 pathogen is easier to

perform than infection with members of MTC which are all classified as BSL 3

pathogens (CHOSEWOOD u. WILSON 2011).

Funding

This project was financially supported by the EMIDA-ERA-NET project

“Mycobactdiagnosis” (grant no. 031A083).

Competing interests

The author(s) declared no potential conflicts of interest with respect to the research,

authorship, and/or publication of this article.

Ethical approval

This study was carried out in strict accordance with European and National Law for

the Care and Use of Animals. The protocol was approved by the Committee on the

Ethics of Animal Experiments and the Protection of Animals of the State of Thuringia,

Germany (Permit Number: 04-002/12).

Page 124: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Manuscript 2

120

Acknowledgement

The authors thank U. Brommer, S. Lied, L. Wirker, L. Wolf for excellent technical

support and W. Maginot for excellent photography.

References (see chapter 9)

Page 125: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

121

6 Discussion

The oral infection of goats with MAH resulted in infections of all inoculated

individuals, different clinical courses of disease and distinct lesions. MAH has been

recognized as subspecies of M. avium since 2002 (MIJS et al. 2002). Infections by

members of M. avium are widely known in livestock species, in particular due to

Johne´s disease in ruminants and tuberculosis in birds (BIET u. BOSCHIROLI 2014).

With the exception of granulomatous lesions of the digestive tract in slaughtered pigs

(HIBIYA et al. 2008; AGDESTEIN et al. 2012), relatively little is known about MAH

infections in other animal species and is primarily based on case reports (HAIST et

al. 2008; KRIZ et al. 2010). The current literature does not report about natural or

experimental infections of goats with MAH (BIET u. BOSCHIROLI 2014). However,

due to the abundance of MAH in the environment (FALKINHAM 2009), it seems

plausible that goats are exposed to this opportunistic pathogen.

In the following: (1) the course of infection, experimental parameters, e.g.

characteristics of the strain of MAH used for inoculation that may have resulted in

severe disease and features of the subclinical infection that may prevent the

recognition will be discussed and (2) the morphological features of the most

prominent lesions induced by MAH, granulomas, will be highlighted.

6.1 Different courses of disease and lesions in goats after oral inoculation with

MAH

The establishment of infection in all MAH inoculated goats demonstrates that goats

are susceptible for MAH although no natural infections have been described so far.

This adds goats to the increasing list of animal species where MAH infections have

been diagnosed (GLAWISCHNIG et al. 2006; HAIST et al. 2008; KRIZ et al. 2010;

AGDESTEIN et al. 2012; KLANG et al. 2014; ARRAZURIA et al. 2015).

One possibility for the severe course of infection observed would be that the strain of

MAH chosen for inoculation was different from circulating strains. Therefore, the MAH

strain 09MA1289 used for inoculation was characterized in detail. It was a porcine

isolate which derived from a slaughtered pig with mycobacteriosis from southern

Germany. The MIRU-VNTR method revealed a genotype profile designated as

Page 126: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

122

22221129 which corresponds to INMV46 (genotypes of MAC members) unique for

Germany until now, but detected before also as porcine MAH isolate described

elsewhere (RADOMSKI et al. 2010). To investigate whether the MAH strain harbors

special virulence features, the presence of different potential virulence markers was

tested, including ISMpa1 and selected genes (JOHANSEN et al. 2009) involved in

the synthesis of GPLs in the cell wall of M. avium (ECKSTEIN et al. 2003;

KRZYWINSKA u. SCHOREY 2003). ISMpa1and the presence of all selected GPLs

synthesis genes distinguishes this MAH strain from those commonly isolated in

Norway (JOHANSEN et al. 2009), but is common in MAH strains from Germany (P.

Möbius, pers. comm.).

For mycobacteria different virulence associated factors are considered to be involved

in the formation of granulomas. For M. tuberculosis, T7SS that secrete proteins

across the cell wall are described (ABDALLAH et al. 2007; STOOP et al. 2012). M.

tuberculosis exhibits 5 different T7SS known as ESX 1-5. ESX-1 was studied in detail

and obtained importance when Mycobacterium bovis BCG was analyzed and it

became clear that the loss of RD1, that encodes ESX-1, was associated with lower

virulence of BCG (ABDALLAH et al. 2007). The genes for ESAT-6 and CFP-10 are

located in RD1. Both proteins are fundamental for the virulence of M. tuberculosis

and the formation of granulomas (STOOP et al. 2012). M. avium species lack the

ESX-1 system (STOOP et al. 2012), nevertheless, multiple granulomas were seen in

this study. This indicates that other factors may drive the process of granuloma

formation independent of the ESX-1 system as recently suggested (STOOP et al.

2012). A possible candidate to address this question could be GPLs that are rich in

the cell wall of M. avium but not in M. tuberculosis (IGNATOV et al. 2012). They are

considered to be highly immunogenic components, capable to regulate the

production of pro- and anti-inflammatory cytokines and are essential in the

establishment of biofilms (JOHANSEN et al. 2009; MUKHERJEE u. CHATTERJI

2012; ROSE u. BERMUDEZ 2014). Even though, the expression of surface GPLs

was not examined in this study, the used MAH strain harbored all genes that encode

for the selected GPLs and thus if GPLs are present they might had a role in

granuloma formation. Although no further data concerning host-pathogen interaction

Page 127: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

123

are available, there is no indication for an increased pathogenicity of this particular

strain.

Another possibility is that the total inoculation dose (2.13 x 1010 cfu/MAH) was

relatively high in comparison with experimental oral MAP infections (HINES II et al.

2007; KRÜGER et al. 2015). The international guidelines for long-term caprine MAP

challenge models recommend the use of 2 consecutive daily doses of 109 organisms

for a successful establishment of infection (HINES II et al. 2007). Thus, size of a

single MAH inoculum was within the aforementioned recommendation, however the

10 times administration resulted in a total dose 5-times as high as recommended and

might have been responsible for the progression towards severe disease in part of

the animals.

After inoculation two different courses of disease were seen. One group developed

severe fatal disease between 2-3 mpi which resulted in unexpected death or even

required euthanasia of goats according to the animal welfare law. All other goats had

a mild transient clinical phase at the same time but remained afterwards healthy with

hidden, persisting lesions detected at necropsy 13 mpi. Inter-individual differences in

the host immune responses are most likely crucial for the course of disease.

Comparable observations were made after infection of goats with MAP in previous

trials. Some animals progressed to severe diffuse multibacillary disease while others

had mild focal paucibacillary lesions (KÖHLER et al. 2015; KRÜGER et al. 2015).

The majority of lesions in the intestine were found in areas with oGALT,

predominantly in the IPP, with extensive ulcerated mucosal surfaces and underlying

granulomatous infiltration. Ulcerative intestinal lesions that are macroscopically

evident are rare in goat kids, however, they shared some features with clostridial

infections (UZAL u. SONGER 2008). Therefore MAH as cause should be considered

when searching for the etiologic agent.

The obvious distribution of lesions almost restricted to oGALT highlights this special

compartment of the intestine as the most likely entry of MAH into the host. This

finding is not astonishing since the follicle-associated epithelium (FAE) overlying

Peyer´s patches is known as port of entry for the closely related MAP (MOMOTANI et

al. 1988; SIGURDARDÓTTIR et al. 2001). M cells are part of the FAE and are

Page 128: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

124

specialized in transcytosis of particulate material from the ingesta as well as

antigens, viruses and bacteria (JUNG et al. 2010). In particular, MAP exploits β1

integrins for entrance by binding via fibronectin which is highly expressed on M cells

(SECOTT et al. 2004; JUNG et al. 2010). However, it should be noted that in a

mouse study invasion of MAH via enterocytes has been found (SANGARI et al.

2001).

In the 9 goats which developed severe disease between 2-3 mpi, different types of

lesions were found in the intestines. One of them had an extensive granulomatous

infiltrate rich in epitheloid cells with many mycobacteria concomitant with small areas

of merely superficial ulceration. The infiltrate was similar to multibacillary lesions seen

in MAP infections of ruminants (CLARKE 1997). Equal lesions were also found in

draining ILN. This might indicate a pathogen-favoring environment which allows

uncontrolled replication of mycobacteria in their target cells.

In 8 of the 9 goats, severely ulcerated intestinal lesions with a lymphocytic dominated

granulomatous infiltrate and scarcely detectable mycobacteria were found. This

extensive tissue damage with massive mucosal ulcerations may have limited

proliferation of MAH as a consequence of an intense local immune response,

however, with detrimental outcome for the host. Severe lesions were also seen in

draining ILN where extensive multifocal to confluent necrosis and mineralization

occurred. Extensive caseous necrosis with circumscribed islands of mineralization

surrounded by a lymphocytic dominated granulomatous infiltrate similar as described

for intestinal lesion was seen, however, with fewer neutrophils. Variable amounts of

mycobacterial material in necrotic areas and single numbers of intracellular

mycobacteria were found by IHC. Since ulcerations cannot occur in ILN, low numbers

of neutrophils might be result of a lack of additionally pro-inflammatory stimuli

induced by gram negative bacteria from the intestinal microflora.

The distribution of lesions and type of inflammatory infiltrate found in 8 of 9 goats

closely resemble those described in bovine tuberculosis when calves were orally

inoculated with Mycobacterium bovis (DOBBERSTEIN u. WILMES 1937; NIEBERLE

1937). In particular the authors reported intestinal lesions restricted to Peyer´s

patches in jejunum and ileum similar as seen in this study. These lesions developed

Page 129: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

125

as classical tubercles, rich in epitheloid cells and MGCs, but M. bovis could also

induce a priori a caseous necrosis of lymphoid tissue which then resulted in ulcerous

eruption (JOEST et al. 1967). Because of the macroscopic resemblance of lesions,

MAH should be considered as differential diagnosis in field cases of intestinal TB in

ruminants (DOBBERSTEIN u. WILMES 1937; NIEBERLE 1937; PESCIAROLI et al.

2014). Finally, it is worth mentioning that in human intestinal TB, ulcerations and

granulomas are commonly seen within the ileocecal region, a site where oGALT is

frequently present (DEBI et al. 2014). This highlights, that species-overlapping and

caused by different mycobacteria similar pathologic lesions may occur.

Bacterial cultivation of MAH revealed a moderate to high bacterial load in tissues of

the digestive tract as well as peripheral organs which indicates a systemic spread

probably due to the blood and lymphatic vessel system. This confirms the histologic

findings of small granulomatous lesions and few granulomas in peripheral organs like

hepatic, retropharyngeal and mediastinal LN. In addition, until 2-3 mpi all inoculated

goats shed large amounts of MAH via feces contaminating the environment.

Especially children could come in close contact with infected animals in zoos and pet

farms (STIRLING et al. 2008). Therefore, a health risk for children, older people and

immunocompromised patients is conceivable (HAIST et al. 2008; KRIZ et al. 2010).

Granulomatous lesions of the digestive tract as consequence of clinically silent

infections with MAH are frequently detected in slaughtered pigs (HIBIYA et al. 2008;

AGDESTEIN et al. 2012; AGDESTEIN et al. 2014). Similar to this, surviving goats

had persisting lesions at the same sites as the goats with severe disease. Multiple

small granulomas were seen in oGALT, predominantly in IPP, and extensive

granulomas that sometimes completely replaced the whole ILN. Histologic

examination revealed granulomas with different morphologies ranging from small

accumulation of epitheloid macrophages to large granulomas with extensive necrosis

and mineralization or thick fibrotic capsules. Morphologically different granulomas are

a common finding in experimental TB in cattle and classification according to cellular

composition and the presence or absence of necrosis, mineralization and fibrosis into

distinct granuloma stages was performed (WANGOO et al. 2005). The scheme from

Wangoo was the basis for staging of granulomas in this thesis. It was modified to

Page 130: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

126

accommodate all histologic granuloma characteristics seen in this study. The

presence of granulomas in longer surviving goats can be interpreted as the ability of

the host`s immune system to limit the inflammation after entry by forming

granulomas. This results in limited intestinal lesions and favors the survival of the

host. However, low amounts of MAH were detectable in few sites of oGALT and ILN

by IHC and bacterial cultivation.

Even though natural infections of goats with MAH are not reported, undetected

infections might be of relevance during slaughter process of goats. Contamination of

meat products during the slaughter process of goats as described for pigs is

conceivable (AGDESTEIN et al. 2014). This assumption is further supported by

recent studies which revealed the presence of MAH by PCR in retail meat from cattle

and pigs (KLANICOVA et al. 2011; KLANICOVA-ZALEWSKA u. SLANA 2014).

6.2 Characterization of morphologic different granuloma stages

Oral infection of goats with MAH resulted in different disease outcomes with the

generation of morphologic and functional different granulomas. To investigate the

cellular composition of granulomas different types of macrophages, T lymphocytes, B

lymphocytes, plasma cells, proliferative and endothelial cells were

immunohistochemically labeled. A consequence of this approach was an expansion

of the granuloma scheme from manuscript 1 involving a unique granuloma stage

seen in ILN at 2-3 mpi.

Histologic differentiation of granuloma morphologies has been performed in mice

(RHOADES et al. 1997), guinea pigs (TURNER et al. 2003) and more frequently in

bovine granulomas of experimental and natural TB (WANGOO et al. 2005; PALMER

et al. 2007; ARANDAY-CORTES et al. 2013; MENIN et al. 2013). The benefit of this

approach is comparability between granulomas and detection of deviations in their

cellular composition induced e.g. by different mycobacterial species (WANGOO et al.

2005). Furthermore, it provides information about the pathogenesis at the local tissue

levels, valuable for testing efficacy of new vaccines and diagnostic methods

(LIEBANA et al. 2007; MENIN et al. 2013). In this study granulomas with different

Page 131: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

127

morphologies were seen in both goat groups and were further analyzed in frozen

tissues from oGALT and ILN.

In brief six stages were finally differentiated: 1-initial small epitheloid macrophage

clusters, 2-solid epitheloid macrophage clusters with minimal necrotic areas, 3-

structured granulomas with monocentric necrosis and mineralization, 4-extensive

multicentric granulomas with necrosis and mineralization surrounded by fibrotic

capsules of varying thickness, 5-fibrotic organization of granulomas with minor

necrosis and mineralization and 6-extensive caseous necrosis without signs of

fibrotic organization.

Many stage 1 granulomas were seen at 2-3 mpi in ILN, whereas few could be

observed in oGALT and ILN at 13 mpi. They were composed of small clusters of

epitheloid macrophages with abundant CD68+ cytoplasm and small numbers of

interspersed PMN and CD4+ T lymphocytes. Similar findings are reported in

experimental bovine TB (WANGOO et al. 2005; PALMER et al. 2007; ARANDAY-

CORTES et al. 2013). In this study, stage 1 granulomas at both time points contained

scattered AFB. Differences in numbers of AFB in stage 1 granulomas were

dependent on the time point of necropsies in experimental bovine TB studies.

Moderate to numerous AFB were detected between 15-60 dpi in experimentally

infected cattle, while at 90 dpi few to none AFB were found (PALMER et al. 2007).

This obvious difference was explained by alternative signaling and activation of

macrophages after the onset of adaptive immunity (PALMER et al. 2007).

Stage 2 granulomas were similarly composed as stage 1, however larger in size and

characterized by minimal areas of predominantly necrotic epitheloid macrophages.

MGCs were first seen at 13 mpi, whereas bovine TB studies reported that MGCs may

have been present as early as 28 dpi (WANGOO et al. 2005; PALMER et al. 2007).

Scattered MAH were detected intracellularly and in necrotic areas and the presence

of DCs indicated antigen presentation with triggering of the adaptive immunity. The

development of mild fibrotic capsules could not be observed, in contrast to findings in

other studies (PALMER et al. 2007; ARANDAY-CORTES et al. 2013).

The areas of minimal necrosis are presumably the origin of more extensive necrosis

seen in stage 3, 4 and 6 granulomas. It remains unclear if stage 6 granulomas

Page 132: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

128

progressed to stage 3 and 4, or if stage 6 granulomas are a unique structure

developed due to an exaggerated immune response. Stage 6 granulomas varied in

size and were found only in ILN of severely sick goats necropsied between 2-3 mpi.

They were composed of extensive caseous necrosis with multiple often confluent

vacuoles and cellular debris. Vacuoles were also reported in a mouse study of

experimental TB and were associated with cholesterol byproducts inside the necrosis

(RHOADES et al. 1997). Under nutrient deficiency mycobacteria can shift their

metabolism towards utilization of cholesterol (EHLERS u. SCHAIBLE 2012) which

might have happened here since many AFB were found inside the necrosis. This

type of granuloma shares some features with transmissive granulomas that are

characterized by growth of mycobacteria, immunopathology and Th1/Th2 dysbalance

(EHLERS u. SCHAIBLE 2012), however neutrophil dominance was not seen at this

stage. A variable zone of cellular debris and degenerated inflammatory cells limiting

the necrosis suggests a dynamic expansion of lesions. The zone of degeneration

was flanked by a granulomatous infiltrate with small clusters of epitheloid

macrophages and dominated by CD4+ T lymphocytes. Experimental mouse studies

revealed a primarily CD4+ T lymphocyte driven immune response, while CD8+ and γδ

T lymphocytes were not involved in host pathogen interaction (APPELBERG et al.

1994; PETROFSKY u. BERMUDEZ 2005). Excessive numbers of CD4+ T

lymphocytes might have secreted abundant IFN-γ, resulting in activation of

macrophages. Activated macrophages secrete TNF-α (DORHOI u. KAUFMANN

2014). Overproduction of TNF-α and IFN- γ results in a hyper-inflammatory milieu

that may lead to necrosis of epitheloid macrophages and inflammatory cells as seen

in the zone of degeneration. Furthermore, mycobacteria were liberated in this

process and may infect newly recruited macrophages. This might lead to new foci of

infections and maintainance of granuloma growth similar as proposed in the

zebrafish model (RAMAKRISHNAN 2012). The exaggerated pro-inflammatory

immune response might also have been responsible for the lack of structured

granulomas and fibrotic capsules, since this would have required a balanced immune

response as seen in latent TB (FLYNN et al. 2011).

Page 133: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

129

The characterization of ulcerative intestinal lesions revealed a CD4+ T lymphocyte

dominated granulomatous inflammation in oGALT. The diffuse CD4+ T lymphocyte

infiltrate with small epitheloid macrophage clusters was similar as in the periphery of

stage 6 granulomas. This excessive pro-inflammatory immune response in the

oGALT might have caused the loss of mucosa and most likely necrotic tissue. This

triggered the influx of neutrophils, which might be further enhanced by invasion of

intestinal bacteria. The diffuse character of this type of lesions was the reason why it

was not considered as granuloma. It can be assumed that the shedding of MAH via

feces is a result of this lesion and therefore transmission into the environment

occurred similar as in open cavitary lesions in human pulmonary TB (BASARABA

2008).

Stage 3, 4 and 5 granulomas were uniquely seen in ILN of healthy goats at 13 mpi. It

has been reported that granulomas with characteristics of stage 3 granulomas

developed as early as 42 dpi in a bovine model of experimental TB infection

(CASSIDY et al. 2001). Stage 3 granulomas were frequently found in oGALT and ILN

and were composed of a central necrosis with mineralization surrounded by a zone

of epitheloid macrophages followed by a zone of lymphocytes. Comparable

observations were made in late stage granulomas of human pulmonary TB, and in

bovine and guinea pig TB (TURNER et al. 2003; ULRICHS et al. 2004; WANGOO et

al. 2005; LIEBANA et al. 2007; PALMER et al. 2007; ARANDAY-CORTES et al.

2013).

A hallmark of stage 3, 4 and 5 granulomas was the spatial organization of different T

and B lymphocyte populations, which progressed with granuloma development. At

stage 3, the zone of lymphocytes differentiated into follicle-like B lymphocyte clusters

and T cell-rich zones. At stage 4 and 5, active germinal centers with light and dark

zones of proliferating cells, surrounded by IFA with DC, many CD4+ T lymphocytes

and decreasing numbers of CD8+, γδ T lymphocytes were found. This spatial

organization of cells fulfills the definition of ectopic lymphoid structures also termed

3rd LT which are seen in chronic infectious diseases and cancer (ULRICHS et al.

2004; PITZALIS et al. 2014). Ectopic lymphoid tissue was recognized in late

granuloma stages of human pulmonary TB (ULRICHS et al. 2004; ULRICHS u.

Page 134: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

130

KAUFMANN 2006). Accumulations of B lymphocytes and arrangement of certain T

lymphocyte populations were reported in experimental and natural infection of cattle

and goats with M. bovis respectively M. caprae (WANGOO et al. 2005; SANCHEZ et

al. 2011; ARANDAY-CORTES et al. 2013). They were rarely interpreted as 3rd LT,

although one study discussed this feature in the context of possible mycobacterial

control due to the local coordinated immune response (ARANDAY-CORTES et al.

2013). The differentiation from pre-existing lymphoid tissue was possible by the

distinct orientation of germinal centers in lymphoid follicles towards the granuloma

center. The relevance of 3rd LT became clear when proliferating B cell clusters

associated with CD68+ antigen presenting cells (APC) harboring mycobacteria were

detected while the central necrosis of granulomas was devoid of them (ULRICHS et

al. 2004; ULRICHS u. KAUFMANN 2006). The authors concluded that mycobacterial

containment is not achieved at the granuloma center, but rather in the 3rd LT where

an intensive crosstalk between lymphocytes and mycobacteria containing APC takes

places resulting in protective immunity. They further questioned the relevance of

cytolytic killing of infected macrophages by CD8+ T lymphocytes and their role for the

development of necrosis since they were primarily found in the outer 3rd LT zone

(ULRICHS et al. 2004; ULRICHS u. KAUFMANN 2006). This finding is further

supported in the guinea pig model of TB (TURNER et al. 2003) and by MAH infection

of goats in this study.

As aforementioned, lymphoid follicles with mycobacteria, might indicate an induction

of humoral immune responses. The role of B lymphocytes in the immune response

against mycobacteria is poorly investigated. However, recent findings suggested that

B lymphocytes can present peptide antigens via MHC-II to prime T lymphocytes,

secrete a variety of cytokines and chemokines and thus might modulate the

granulomatous inflammation and influence the control of TB infection (FLYNN et al.

2011; CHAN et al. 2014; HOFF et al. 2015). Besides possible antigen presentation in

lymphoid follicles other compartments might be of relevance. The presence of DCs in

IFA, in the granulomatous layer and near necrosis may also have contributed to a

successful immune response since these highly mobile antigen presenting cells

Page 135: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

131

might have migrated from the granuloma center to the 3rd LT where they activated T

lymphocytes.

The zone of connective tissue separated granulomatous infiltrate from 3rd LT and was

a trait of stage 4 and 5 granulomas, but not of stage 3 granulomas. Similar findings

have been reported in late stage granulomas of human pulmonary TB (ULRICHS et

al. 2004). This is different from the complete fibrous capsules which surround bovine

stage 3 granulomas (WANGOO et al. 2005; ARANDAY-CORTES et al. 2013). It is

very likely that the extensive necrosis with mineralization and moderate fibrosis of

stage 4, progressed to fibrotic organization which encircled mineralized foci of stage

5 granulomas. It has been stated that the progress of granulomas towards

mineralization of necrosis together with fibrosis can be viewed as healing process

beneficial for the host (BASARABA 2008).

Labeling with the proliferation marker Ki-67 revealed foci of proliferative cells within

germinal centers of 3rd LT. Some proliferative cells were found inside the granulomas

which indicate limited local expansion of inflammatory cells. When comparing the

vascularization of different granuloma zones, it seems plausible that the highly

vascularized granulomatous zone of stage 3 granulomas promotes influx of

inflammatory cells. The vascularization of stage 3 granulomas was similar to

tuberculomas described in human pulmonary TB (ULRICHS et al. 2005). Ulrichs and

colleagues concluded that highly vascularized tuberculomas allowed oxygen and

nutrient supply necessary for a productive crosstalk between the host and M.

tuberculosis (ULRICHS et al. 2005). The 3rd LT was similarly vascularized as regular

lymphoid tissue. Reduced vascularization was seen in granulomatous infiltrates and

was almost absent in zones of connective tissue in stage 4 and 5. In granulomas of

guinea pig models of TB similar findings were reported with fibrotic organization

resulting in compression of vessels near the necrotic granuloma core (ORME u.

BASARABA 2014). The consequence of this process was low oxygen supply and

hypoxia within tissue (VIA et al. 2008; ORME u. BASARABA 2014). A hypoxia

marker was not included in the assessment of granulomas in this study, however, it

seems very likely that hypoxia also occurs in these structures. In summary, low

numbers of AFB and well advanced mineralization and fibrosis with reduced

Page 136: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

132

vascularization, has been associated with sterilized and healed granulomas

(BASARABA 2008).

6.3 Outlook and possible directions of further research

The experimental oral infection of goats with MAH may be a valuable model for

comparative TB research in several aspects:

The different courses of diseases allow the examination of certain TB entities, e.g.

granulomatous lesions seen in intestinal TB or granulomas seen in latent TB.

Especially, granulomas seen in the surviving goats resemble the heterogeneity of

lesions reported in cattle and goats after infection with MTC members, as well as

lesions in humans with pulmonary TB (VAN RHIJN et al. 2008; LENAERTS et al.

2015). The development of 3rd LT with well-organized granulomas has been reported

in few studies only (ULRICHS et al. 2004; ULRICHS et al. 2005; ARANDAY-

CORTES et al. 2013). Therefore, the presence of 3rd LT is a major advantage of this

model to evaluate the relevance of this feature in the control of mycobacteria by the

host immune response. Drug efficacy testing is another aspect for experimental TB

models. The plethora of morphologically different granuloma stages could be of great

interest for drug testing. Certain features like necrosis, mineralization, extensive

fibrosis and vascularization of granulomas directly influence the pharmacodynamics

and pharmacokinetics of administered drugs and might be evaluable by

histopathology and modern live imaging in this goat model (BASARABA 2008;

BARRY et al. 2009).

The relative small size of goats allows much lower housing and feeding costs when

compared with cattle (GONZALEZ-JUARRERO et al. 2013). In comparison to other

livestock species, the goat model allows the utilization of modern live imaging

systems, e.g. magnetic resonance imaging, computed tomography or positron

emission tomography (DE VAL et al. 2011; LENAERTS et al. 2015). This feature

could be of special value to monitor granuloma development in experimentally

infected goats. The lack of a wide array of immunologic tools is usually considered as

a constraint of large animal models (YOUNG 2009). However, the recent

development of novel in-situ hybridization (ISH) methods might overcome this

Page 137: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Discussion

133

obstacle as indicated recently by cytokine gene expression analysis in bovine

pulmonary TB granulomas (PALMER et al. 2015).

Finally, another benefit of the experimental design was the classification of MAH as

BSL 2 pathogen allowing an easier experimental performance than BSL 3 condition

would have required. Since all MTC members are BSL 3 pathogens, special

requirements for housing, occupational safety and tissue processing have to be

considered (CHOSEWOOD u. WILSON 2011).

In conclusion, the obvious similarities of granulomas with those seen in bovine and

human TB and the association of ectopic lymphoid tissue with organized granulomas

highlights this model as appropriate for comparative TB and NTM research and the

goat in particular as model species.

Page 138: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Summary

134

7 Summary

Jan Schinköthe:

Experimental oral infection of goats with Mycobacterium avium subsp.

hominissuis: Pathomorphological characterization of lesions in different

courses of disease with special focus on cellular composition of granulomas

Goats were experimentally infected with Mycobacterium avium subsp. hominissuis

(MAH) to determine the specificity of diagnostic antigens for Mycobacterium avium

subsp. paratuberculosis. For this, each of 21 goats was inoculated orally 10-times,

every 2-4 days with MAH receiving a total dose of 2.13 x 1010 cfu MAH. An

unexpected onset of clinical signs progressing to severe disease was observed in 50

% of the goats at 2-3 mpi. The remaining goats had mild transient clinical signs and

were healthy 13 mpi at the end of the trial. The first objective of this study was to

characterize lesions in severely sick goats and in surviving goats at 13 mpi and to

examine lesions for presence of MAH.

At necropsy, goats were examined for macroscopic lesions. Representative samples

of lesions, intestinal tissues, lymphoid organs and large parenchyma were fixed in

NBF and snap frozen. Histologic lesions were evaluated in HE stained FFPE

sections. Detection of MAH in sampled tissues was performed by IHC and by

bacterial cultivation.

Goats necropsied at 2-3 mpi had severe intestinal ulcerations with underlying

granulomatous infiltrates in oGALT and large granulomas in ILN with variable

quantities of MAH detected in tissue sections. Large amounts of MAH were cultivated

from intestinal and ILN lesions and peripheral organs, indicating extra-intestinal

spread of MAH. At 13 mpi, multiple small granulomas in oGALT and large

granulomas in ILN were seen at necropsy. MAH was detected by IHC in few

granulomas (3/9 goats) and cultured in low quantities in 6/9 goats at intestinal sites

and ILN only. All goats excreted moderate to high amounts of MAH via feces until 2

mpi. Environmental contamination due to fecal shedding and low amounts of

Page 139: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Summary

135

mycobacteria in granulomas of otherwise healthy goats may represent a potential

risk for human health.

Since large numbers of morphologically heterogeneous granulomas were found in

goats at 2-3 mpi and 13 mpi, the second objective was to characterize their cellular

composition and to compare them with granulomas seen in human and bovine

tuberculosis. In consecutive frozen sections, CD4+, CD8+, γδ T lymphocytes, B

lymphocytes, plasma cells, CD68+, CD25+, MHC-II+, proliferative and endothelial cells

were labeled by IHC. Furthermore, collagen and AFB were stained by Azan,

respectively, Kinyoun method.

Granulomas with extensive necrosis, little mineralization and variable numbers of

AFB surrounded by many CD4+ T cells, but only few epitheloid macrophages were

observed in severely sick goats at 2-3 mpi. They were interpreted as exuberant

immune reaction. Organized granulomas with very few AFB were seen in clinically

healthy goats at 13 mpi. The necrotic cores were surrounded by a zone of

granulomatous infiltrate with many epitheloid macrophages and few lymphocytes.

This zone was initially wide and highly vascularized and became progressively

smaller. It was enclosed by an increasing layer of connective tissue. All organized

granulomas were surrounded by compartimentalized tertiary lymphoid tissue. The

granulomas in experimental infection of goats with MAH reflect the heterogeneity of

lesions seen in mycobacterial infections of humans and ruminants.

In conclusion, the experimental infection of goats with MAH resulted in different

courses of disease, morphologically different granulomas with striking similarities to

those seen in bovine and human TB, and the development of ectopic lymphoid tissue

in organized granulomas. These features highlight this model as appropriate for

comparative TB and NTM research and the goat in particular as model species.

Page 140: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Zusammenfassung

136

8 Zusammenfassung

Jan Schinköthe:

Experimentelle orale Infektion von Ziegen mit Mycobacterium avium subsp.

hominissuis: Pathomorphologische Charakterisierung der Läsionen bei

verschiedenen Krankheitsverläufen unter besonderer Berücksichtigung der

zellulären Zusammensetzung der Granulome

Ziegen wurden experimentell mit Mycobacterium avium subsp. hominissuis (MAH)

infiziert, um die Spezifität von diagnostischen Antigenen gegen Mycobacterium

avium subsp. paratuberculosis zu testen. Hierfür wurde eine Gruppe von 21 Ziegen

10-mal, alle 2-4 Tage mit einer Gesamtdosis von 2,13 x 1010 KBE MAH oral

inokuliert. Unerwartet traten bei 50% der Ziegen 2-3 Monate nach Erstinfektion (Mpi)

schwere klinische Symptome auf und mehrere Tiere verendeten spontan oder

mussten aus ethischen Gründen getötet werden. Die überlebenden Ziegen zeigten

milde transiente klinische Symptome und waren bei der Sektion 13 Mpi klinisch

gesund. Das erste Ziel dieser Studie war, die Läsionen sowohl bei den hochgradig

Erkrankten als auch bei den überlebenden Ziegen zu charakterisieren und auf das

Vorhandensein von MAH zu untersuchen.

Bei der Sektion wurden die makroskopischen Läsionen dokumentiert und

repräsentative Proben von den Läsionen, intestinalen Geweben sowie lymphatischen

und peripheren Organen entnommen und in Formalin fixiert bzw. kryokonserviert. Die

histopathologische Analyse der Läsionen fand an HE-gefärbten Paraffinschnitten

statt. Der Nachweis von MAH wurde mit Immunhistochemie (IHC) am Schnitt und mit

bakterieller Kultivierung durchgeführt.

Ziegen zum Zeitpunkt 2-3 Mpi wiesen hochgradige intestinale Ulzerationen mit

zugrunde liegenden granulomatösen Infiltraten in organisiertem Darm-assoziierten

lymphatischen Gewebe (oGALT) und große Granulome in intestinalen Lymphknoten

(ILN) auf. MAH konnte mit IHC in variabler Zahl intra- und extrazellulär nachgewiesen

werden. Eine hohe Erregerlast wurde in intestinalen und ILN Läsionen sowie in

peripheren Organen durch bakterielle Kultur festgestellt. Dies deutet auf eine

Page 141: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Zusammenfassung

137

extraintestinale Verbreitung von MAH hin. 13 Mpi wurden bei allen untersuchten

Ziegen mehrere kleine Granulome in oGALT und/oder große Granulome in ILN bei

der Sektion gefunden. MAH ließ sich nur in wenigen Granulomen (3/9 Ziegen) durch

IHC nachweisen bzw. aus 6/9 Ziegen in geringen Mengen kultivieren. Alle infizierten

Ziegen schieden bis 2 Mpi moderate bis hohe Mengen an MAH über den Kot aus.

Die dabei eintretende Umgebungskontamination sowie das Vorhandensein von

Mykobakterien in Granulomen von gesunden Ziegen könnte ein mögliches Risiko für

die menschliche Gesundheit darstellen.

Da in den Ziegen 2-3 und 13 Mpi eine große Anzahl morphologisch heterogener

Granulome gefunden wurde, war das zweite Ziel dieser Studie herauszufinden, wie

die zelluläre Zusammensetzung der Granulome ist und diese mit denen bei der

Tuberkulose der Rindes und des Menschen zu vergleichen. Dafür wurden in

Konsekutivschnitten CD4+, CD8+, γδ-T-Lymphozyten, B-Lymphozyten, Plasmazellen,

CD68+, CD25+, MHC-II+, proliferative Zellen und Endothelzellen mittels IHC markiert.

Kollagen und säurefeste Bakterien (AFB) wurden mit Azan- bzw. Kinyounfärbung

dargestellt.

Granulome mit extensiven verkäsenden Nekrosen, die minimale Mineralisierungen

und viele AFB enthielten, waren umgeben von vielen CD4+ T Lymphozyten mit

wenigen epitheloiden Makrophagen und waren nur in ILN 2-3 Mpi vorhanden. Diese

Befunde wurden als überschießende Immunantwort interpretiert. Organisierte

Granulome mit sehr wenigen AFB wurden in klinisch gesunden Ziegen 13 Mpi

gefunden. Die nekrotischen Zentren dieser Granulome waren umgeben von einer

Zone aus granulomatösem Infiltrat mit vielen epitheloiden Makrophagen und wenigen

Lymphozyten. Diese Zone war zu Beginn breit und gut vaskularisiert. Sie wurde

fortschreitend schmaler und stattdessen entwickelte sich eine immer breiter

werdende Schicht aus Bindegewebe. Alle organisierten Granulome waren zusätzlich

von organisiertem tertiärem lymphatischem Gewebe umgeben. Die Heterogenität der

Granulome bei MAH infizierten Ziegen zeigte eine große Ähnlichkeit mit Granulomen

der Tuberkulose des Rindes und des Menschen.

Die verschiedenen Krankheitsverläufe mit Latenzphase, das Auftreten von

morphologisch heterogenen Granulomen und die Entwicklung von tertiärem

Page 142: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Zusammenfassung

138

lymphatischem Gewebe in organisierten Granulomen machen die experimentelle

Infektion der Ziege mit MAH zu einem für die vergleichende Forschung an

Infektionen mit tuberkulösen und nicht tuberkulösen Mykobakterien hervorragend

geeigneten Modell.

Page 143: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

139

9 References

ABDALLAH, A. M., N. C. GEY VAN PITTIUS, P. A. CHAMPION, J. COX, J. LUIRINK, C. M. VANDENBROUCKE-GRAULS, B. J. APPELMELK u. W. BITTER (2007): Type VII secretion--mycobacteria show the way. Nat. Rev. Microbiol. 5, 883-891 AGDESTEIN, A., T. B. JOHANSEN, O. KOLBJORNSEN, A. JORGENSEN, B. DJONNE u. I. OLSEN (2012): A comparative study of Mycobacterium avium subsp. avium and Mycobacterium avium subsp. hominissuis in experimentally infected pigs. BMC Vet. Res. 8, 11 AGDESTEIN, A., I. OLSEN, A. JØRGENSEN, B. DJØNNE u. T. B. JOHANSEN (2014): Novel insights into transmission routes of Mycobacterium avium in pigs and possible implications for human health. Vet. Res. 45, 46 AMENI, G., M. VORDERMEIER, R. FIRDESSA, A. ASEFFA, G. HEWINSON, S. V. GORDON u. S. BERG (2011): Mycobacterium tuberculosis infection in grazing cattle in central Ethiopia. The Veterinary Journal 188, 359-361 APPELBERG, R., A. G. CASTRO, J. PEDROSA, R. A. SILVA, I. M. ORME u. P. MINÓPRIO (1994): Role of gamma interferon and tumor necrosis factor alpha during T-cell-independent and-dependent phases of Mycobacterium avium infection. Infect. Immun. 62, 3962-3971 ARANDAY-CORTES, E., N. C. BULL, B. VILLARREAL-RAMOS, J. GOUGH, D. HICKS, Á. ORTIZ-PELÁEZ, H. M. VORDERMEIER u. F. J. SALGUERO (2013): Upregulation of IL-17A, CXCL9 and CXCL10 in Early-Stage Granulomas Induced by Mycobacterium bovis in Cattle. Transboundary and Emerging Diseases 60, 525-537 ARIS, F., C. NAIM, T. BESSISSOW, R. AMRE u. G. P. ARTHO (2011): AIRP best cases in radiologic-pathologic correlation: Mycobacterium avium-intracellulare complex enteritis. Radiographics 31, 825-830 ARRAZURIA, R., I. A. SEVILLA, E. MOLINA, V. PEREZ, J. M. GARRIDO, R. A. JUSTE u. N. ELGUEZABAL (2015): Detection of Mycobacterium avium subspecies in the gut associated lymphoid tissue of slaughtered rabbits. BMC Vet. Res. 11, 130

Page 144: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

140

BARRY, C. E., H. I. BOSHOFF, V. DARTOIS, T. DICK, S. EHRT, J. FLYNN, D. SCHNAPPINGER, R. J. WILKINSON u. D. YOUNG (2009): The spectrum of latent tuberculosis: rethinking the biology and intervention strategies. Nature Reviews Microbiology 7, 845-855 BASARABA, R. J. (2008): Experimental tuberculosis: the role of comparative pathology in the discovery of improved tuberculosis treatment strategies. Tuberculosis (Edinb) 88 Suppl 1, S35-47 BEHAR, S. M., M. DIVANGAHI u. H. G. REMOLD (2010): Evasion of innate immunity by Mycobacterium tuberculosis: is death an exit strategy? Nature Reviews Microbiology 8, 668-674 BEHR, M. A. u. J. O. FALKINHAM III (2009): Molecular epidemiology of nontuberculous mycobacteria. Future Microbiol. 4, 1009-1020 BENINI, J., E. M. EHLERS u. S. EHLERS (1999):

Different types of pulmonary granuloma necrosis in immunocompetent vs. TNFRp55‐gene‐deficient mice aerogenically infected with highly virulent Mycobacterium avium. The Journal of pathology 189, 127-137 BERMUDEZ, L. E., M. PETROFSKY, P. KOLONOSKI u. L. S. YOUNG (1992): An Animal Model of Mycobacterium avium Complex Disseminated Infection after Colonization of the Intestinal Tract. J. Infect. Dis. 165, 75-79 BHATNAGAR, S., K. SHINAGAWA, F. J. CASTELLINO u. J. S. SCHOREY (2007): Exosomes released from macrophages infected with intracellular pathogens stimulate a proinflammatory response in vitro and in vivo. Blood 110, 3234-3244 BIET, F. u. M. L. BOSCHIROLI (2014): Non-tuberculous mycobacterial infections of veterinary relevance. Res. Vet. Sci. 97, Supplement, S69-S77 BIET, F., M. L. BOSCHIROLI, M. F. THOREL u. L. A. GUILLOTEAU (2005): Zoonotic aspects of Mycobacterium bovis and Mycobacterium avium-intracellulare complex (MAC). Vet. Res. 36, 411-436 BLOMGRAN, R. u. J. D. ERNST (2011): Lung neutrophils facilitate activation of naive antigen-specific CD4+ T cells during Mycobacterium tuberculosis infection. The Journal of Immunology 186, 7110-7119

Page 145: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

141

BMEL (2015): Meldepflichtige Tierkrankheiten. BRENNAN, P. J. (2003): Structure, function, and biogenesis of the cell wall of Mycobacterium tuberculosis. Tuberculosis 83, 91-97 BRODE, S. K., C. L. DALEY u. T. K. MARRAS (2014): The epidemiologic relationship between tuberculosis and non-tuberculous mycobacterial disease: a systematic review. Int. J. Tuberc. Lung Dis. 18, 1370-1377 BRUIJNESTEIJN VAN COPPENRAET, L. E., P. E. DE HAAS, J. A. LINDEBOOM, E. J. KUIJPER u. D. VAN SOOLINGEN (2008): Lymphadenitis in children is caused by Mycobacterium avium hominissuis and not related to 'bird tuberculosis'. Eur. J. Clin. Microbiol. Infect. Dis. 27, 293-299 CAPUANO, S. V., D. A. CROIX, S. PAWAR, A. ZINOVIK, A. MYERS, P. L. LIN, S. BISSEL, C. FUHRMAN, E. KLEIN u. J. L. FLYNN (2003): Experimental Mycobacterium tuberculosis infection of cynomolgus macaques closely resembles the various manifestations of human M. tuberculosis infection. Infect. Immun. 71, 5831-5844 CARDONA, P.-J. u. J. IVANYI (2011): The secret trumps, impelling the pathogenicity of tubercle bacilli. Enferm. Infecc. Microbiol. Clin. 29, 14-19 CARDONA, P. J. (2015): The key role of exudative lesions and their encapsulation: lessons learned from the pathology of human pulmonary tuberculosis. Frontiers in microbiology 6, 612 CASSIDY, J., D. BRYSON, M. G. CANCELA, F. FORSTER, J. POLLOCK u. S. NEILL (2001): Lymphocyte subtypes in experimentally induced early-stage bovine tuberculous lesions. J. Comp. Pathol. 124, 46-51 CHAN, J., S. MEHTA, S. BHARRHAN, Y. CHEN, J. M. ACHKAR, A. CASADEVALL u. J. FLYNN (2014): The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. Semin. Immunol. 26, 588-600

Page 146: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

142

CHOSEWOOD, L. u. D. WILSON (2011): Biosafety in Microbiological and Biomedical Laboratories (US Department of Health and Human Services, Washington, DC). Availableat http://www. cdc. gov/biosafety/publications/bmbl5/BMBL. pdf. Accessed 08.08.2015 CLARKE, C. J. (1997): The pathology and pathogenesis of paratuberculosis in ruminants and other species. J. Comp. Pathol. 116, 217-261 CRAWSHAW, T., R. DANIEL, R. CLIFTON-HADLEY, J. CLARK, H. EVANS, S. ROLFE u. R. DE LA RUA-DOMENECH (2008): TB in goats caused by Mycobacterium bovis. Vet. Rec. 163, 127 DAMSKER, B. u. E. J. BOTTONE (1985): Mycobacterium avium-Mycobacterium intracellulare from the intestinal tracts of patients with the acquired immunodeficiency syndrome: concepts regarding acquisition and pathogenesis. The Journal of infectious diseases 179-181 DAVIS, J. M. u. L. RAMAKRISHNAN (2009): The Role of the Granuloma in Expansion and Dissemination of Early Tuberculous Infection. Cell 136, 37-49 DE VAL, B. P., S. LÓPEZ-SORIA, M. NOFRARÍAS, M. MARTÍN, H. M. VORDERMEIER, B. VILLARREAL-RAMOS, N. ROMERA, M. ESCOBAR, D. SOLANES u. P.-J. CARDONA (2011): Experimental model of tuberculosis in the domestic goat after endobronchial infection with Mycobacterium caprae. Clinical and Vaccine Immunology 18, 1872-1881 DE VAL PEREZ, B., S. LOPEZ-SORIA, M. NOFRARIAS, M. MARTIN, H. M. VORDERMEIER, B. VILLARREAL-RAMOS, N. ROMERA, M. ESCOBAR, D. SOLANES, P. J. CARDONA u. M. DOMINGO (2011): Experimental model of tuberculosis in the domestic goat after endobronchial infection with Mycobacterium caprae. Clin. Vaccine Immunol. 18, 1872-1881 DEBI, U., V. RAVISANKAR, K. K. PRASAD, S. K. SINHA u. A. K. SHARMA (2014): Abdominal tuberculosis of the gastrointestinal tract: revisited. World J. Gastroenterol. 20, 14831-14840

Page 147: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

143

DESPIERRES, L., S. COHEN-BACRIE, H. RICHET u. M. DRANCOURT (2012): Diversity of Mycobacterium avium subsp. hominissuis mycobacteria causing lymphadenitis, France. European Journal of Clinical Microbiology & Infectious Diseases 31, 1373-1379 DHAMA, K., M. MAHENDRAN, R. TIWARI, S. DAYAL SINGH, D. KUMAR, S. SINGH u. P. M. SAWANT (2011): Tuberculosis in birds: Insights into the Mycobacterium avium infections. Veterinary medicine international 2011, DIVANGAHI, M., D. DESJARDINS, C. NUNES-ALVES, H. G. REMOLD u. S. M. BEHAR (2010): Eicosanoid pathways regulate adaptive immunity to Mycobacterium tuberculosis. Nat. Immunol. 11, 751-758 DOBBERSTEIN, J. u. H. WILMES (1937): Zur Pathogenese und Histogenese der experimentellen Fütterungstuberkulose des Kalbes nebst Beiträgen zu ihrer Altersbestimmung. Arch. wiss. prakt. Tierhlk. 71, p. 427 DOMINGO, M., O. GIL, E. SERRANO, E. GUIRADO, M. NOFRARIAS, M. GRASSA, N. CACERES, B. PEREZ, C. VILAPLANA u. P. J. CARDONA (2009): Effectiveness and safety of a treatment regimen based on isoniazid plus vaccination with Mycobacterium tuberculosis cells' fragments: field-study with naturally Mycobacterium caprae-infected goats. Scand. J. Immunol. 69, 500-507 DOMINGO, M., E. VIDAL u. A. MARCO (2014): Pathology of bovine tuberculosis. Res. Vet. Sci. 97, Supplement, S20-S29 DORHOI, A. u. S. H. E. KAUFMANN (2014): Tumor necrosis factor alpha in mycobacterial infection. Semin. Immunol. 26, 203-209 DRIVER, E. R., G. J. RYAN, D. R. HOFF, S. M. IRWIN, R. J. BASARABA, I. KRAMNIK u. A. J. LENAERTS (2012): Evaluation of a mouse model of necrotic granuloma formation using C3HeB/FeJ mice for testing of drugs against Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 56, 3181-3195 ECKSTEIN, T. M., J. T. BELISLE u. J. M. INAMINE (2003): Proposed pathway for the biosynthesis of serovar-specific glycopeptidolipids in Mycobacterium avium serovar 2. Microbiology 149, 2797-2807

Page 148: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

144

EHLERS, S. u. U. E. SCHAIBLE (2012): The granuloma in tuberculosis: dynamics of a host-pathogen collusion. Front Immunol 3, 411 ELZE, J., E. LIEBLER-TENORIO, M. ZILLER u. H. KOHLER (2013): Comparison of prevalence estimation of Mycobacterium avium subsp. paratuberculosis infection by sampling slaughtered cattle with macroscopic lesions vs. systematic sampling. Epidemiol. Infect. 141, 1536-1544 ENG, J. W.-L., C. B. REED, K. M. KOKOLUS, R. PITONIAK, A. UTLEY, M. J. BUCSEK, W. W. MA, E. A. REPASKY u. B. L. HYLANDER (2015): Housing temperature-induced stress drives therapeutic resistance in murine tumour models through β2-adrenergic receptor activation. Nature communications 6, ENGLUND, S., A. BALLAGI-PORDANY, G. BOLSKE u. K. E. JOHANSSON (1999): Single PCR and nested PCR with a mimic molecule for detection of Mycobacterium avium subsp. paratuberculosis. Diagn. Microbiol. Infect. Dis. 33, 163-171 FALKINHAM, J. O., 3RD (2009): Surrounded by mycobacteria: nontuberculous mycobacteria in the human environment. J. Appl. Microbiol. 107, 356-367 FALKINHAM, J. O., 3RD (2013): Reducing human exposure to Mycobacterium avium. Ann Am Thorac Soc 10, 378-382 FATTORINI, L., M. MATTEI, R. PLACIDO, B. LI, E. IONA, U. AGRIMI, V. COLIZZI u. G. OREFICI (1999): Mycobacterium avium infection in BALB/c and SCID mice. J. Med. Microbiol. 48, 577-583 FLORIDO, M., A. M. COOPER u. R. APPELBERG (2002): Immunological basis of the development of necrotic lesions following Mycobacterium avium infection. Immunology 106, 590-601 FLYNN, J. L., J. CHAN u. P. L. LIN (2011): Macrophages and control of granulomatous inflammation in tuberculosis. Mucosal Immunol 4, 271-278 FRITSCH, I., G. LUYVEN, H. KÖHLER, W. LUTZ u. P. MÖBIUS (2012): Suspicion of Mycobacterium avium subsp. paratuberculosis transmission between cattle and wild-living red deer (Cervus elaphus) by multitarget genotyping. Appl. Environ. Microbiol. 78, 1132-1139

Page 149: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

145

GARCÍA-JIMÉNEZ, W. L., J. M. BENÍTEZ-MEDINA, R. MARTÍNEZ, J. CARRANZA, R. CERRATO, A. GARCÍA-SÁNCHEZ, D. RISCO, J. C. MORENO, M. SEQUEDA, L. GÓMEZ, P. FERNÁNDEZ-LLARIO u. J. HERMOSO-DE-MENDOZA (2015): Non-tuberculous Mycobacteria in Wild Boar (Sus scrofa) from Southern Spain: Epidemiological, Clinical and Diagnostic Concerns. Transboundary and Emerging Diseases 62, 72-80 GIL, O., I. DÍAZ, C. VILAPLANA, G. TAPIA, J. DÍAZ, M. FORT, N. CÁCERES, S. PINTO, J. CAYLÀ u. L. CORNER (2010): Granuloma encapsulation is a key factor for containing tuberculosis infection in minipigs. PLoS ONE 5, e10030 GLAWISCHNIG, W., T. STEINECK u. J. SPERGSER (2006): Infections caused by Mycobacterium avium subspecies avium, hominissuis, and paratuberculosis in free-ranging red deer (Cervus elaphus hippelaphus) in Austria, 2001-2004. J. Wildl. Dis. 42, 724-731 GONZALEZ-JUARRERO, M., A. BOSCO-LAUTH, B. PODELL, C. SOFFLER, E. BROOKS, A. IZZO, J. SANCHEZ-CAMPILLO u. R. BOWEN (2013): Experimental aerosol Mycobacterium bovis model of infection in goats. Tuberculosis 93, 558-564 GRIFFITH, D. E., T. AKSAMIT, B. A. BROWN-ELLIOTT, A. CATANZARO, C. DALEY, F. GORDIN, S. M. HOLLAND, R. HORSBURGH, G. HUITT, M. F. IADEMARCO, M. ISEMAN, K. OLIVIER, S. RUOSS, C. F. VON REYN, R. J. WALLACE u. K. WINTHROP (2007): An Official ATS/IDSA Statement: Diagnosis, Treatment, and Prevention of Nontuberculous Mycobacterial Diseases. Am. J. Respir. Crit. Care Med. 175, 367-416 GUERRERO, C., C. BERNASCONI, D. BURKI, T. BODMER u. A. TELENTI (1995): A novel insertion element from Mycobacterium avium, IS1245, is a specific target for analysis of strain relatedness. J. Clin. Microbiol. 33, 304-307 GUIRADO, E. u. L. S. SCHLESINGER (2013): Modeling the Mycobacterium tuberculosis Granuloma – the Critical Battlefield in Host Immunity and Disease. Frontiers in Immunology 4, 98 HAIST, V., F. SEEHUSEN, I. MOSER, H. HOTZEL, U. DESCHL, W. BAUMGARTNER u. P. WOHLSEIN (2008): Mycobacterium avium subsp. hominissuis infection in 2 pet dogs, Germany. Emerg. Infect. Dis. 14, 988-990

Page 150: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

146

HAUG, M., J. A. AWUH, M. STEIGEDAL, J. FRENGEN KOJEN, A. MARSTAD, I. S. NORDRUM, Ø. HALAAS u. T. H. FLO (2013): Dynamics of immune effector mechanisms during infection with Mycobacterium avium in C57BL/6 mice. Immunology 140, 232-243 HIBIYA, K., M. FURUGEN, F. HIGA, M. TATEYAMA u. J. FUJITA (2011): Pigs as an experimental model for systemic Mycobacterium avium infectious disease. Comp. Immunol. Microbiol. Infect. Dis. 34, 455-464 HIBIYA, K., Y. KASUMI, I. SUGAWARA u. J. FUJITA (2008): Histopathological classification of systemic Mycobacterium avium complex infections in slaughtered domestic pigs. Comp. Immunol. Microbiol. Infect. Dis. 31, 347-366 HIBIYA, K., K. UTSUNOMIYA, T. YOSHIDA, S. TOMA, F. HIGA, M. TATEYAMA u. J. FUJITA (2010): Pathogenesis of systemic Mycobacterium avium infection in pigs through histological analysis of hepatic lesions. Can. J. Vet. Res. 74, 252-257 HINES II, M. E., J. R. STABEL, R. W. SWEENEY, F. GRIFFIN, A. M. TALAAT, D. BAKKER, G. BENEDICTUS, W. C. DAVIS, G. W. DE LISLE, I. A. GARDNER, R. A. JUSTE, V. KAPUR, A. KOETS, J. MCNAIR, G. PRUITT u. R. H. WHITLOCK (2007): Experimental challenge models for Johne's disease: A review and proposed international guidelines. Vet. Microbiol. 122, 197-222 HOEFSLOOT, W., J. VAN INGEN, C. ANDREJAK, K. ÄNGEBY, R. BAURIAUD, P. BEMER, N. BEYLIS, M. J. BOEREE, J. CACHO u. V. CHIHOTA (2013): The geographic diversity of nontuberculous mycobacteria isolated from pulmonary samples: an NTM-NET collaborative study. Eur. Respir. J. 42, 1604-1613 HOFF, S. T., A. M. SALMAN, M. RUHWALD, P. RAVN, I. BROCK, N. ELSHEIKH, P. ANDERSEN u. E. M. AGGER (2015): Human B cells produce chemokine CXCL10 in the presence of Mycobacterium tuberculosis specific T cells. Tuberculosis 95, 40-47 HONDA, J. R., V. KNIGHT u. E. D. CHAN (2015): Pathogenesis and risk factors for nontuberculous mycobacterial lung disease. Clin. Chest Med. 36, 1-11 IGNATOV, D., E. KONDRATIEVA, T. AZHIKINA u. A. APT (2012): Mycobacterium avium-triggered diseases: pathogenomics. Cell. Microbiol. 14, 808-818

Page 151: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

147

JOEST, E., J. DOBBERSTEIN, G. PALLASKE u. H. STUNZI (1967): Handbuch der speziellen pathologischen Anatomie der Haustiere. P. Parey, JOHANSEN, T. B., A. AGDESTEIN, I. OLSEN, S. F. NILSEN, G. HOLSTAD u. B. DJONNE (2009): Biofilm formation by Mycobacterium avium isolates originating from humans, swine and birds. BMC Microbiol. 9, 159 JOHANSEN, T. B., B. DJONNE, M. R. JENSEN u. I. OLSEN (2005): Distribution of IS1311 and IS1245 in Mycobacterium avium subspecies revisited. J. Clin. Microbiol. 43, 2500-2502 JUNG, C., J. P. HUGOT u. F. BARREAU (2010): Peyer's Patches: The Immune Sensors of the Intestine. Int J Inflam 2010, 823710 KARP, C. L. (2012): Unstressing intemperate models: how cold stress undermines mouse modeling. The Journal of experimental medicine 209, 1069-1074 KARTALIJA, M., A. R. OVRUTSKY, C. L. BRYAN, G. B. POTT, G. FANTUZZI, J. THOMAS, M. J. STRAND, X. BAI, P. RAMAMOORTHY, M. S. ROTHMAN, V. NAGABHUSHANAM, M. MCDERMOTT, A. R. LEVIN, A. FRAZER-ABEL, P. C. GICLAS, J. KORNER, M. D. ISEMAN, L. SHAPIRO u. E. D. CHAN (2013): Patients with Nontuberculous Mycobacterial Lung Disease Exhibit Unique Body and Immune Phenotypes. Am. J. Respir. Crit. Care Med. 187, 197-205 KASSA, G. M., F. ABEBE, Y. WORKU, M. LEGESSE, G. MEDHIN, G. BJUNE u. G. AMENI (2012): Tuberculosis in goats and sheep in afar pastoral region of ethiopia and isolation of Mycobacterium tuberculosis from goat. Veterinary medicine international 2012, KAUFMANN, S. H. E. u. A. DORHOI (2013): Inflammation in tuberculosis: interactions, imbalances and interventions. Curr. Opin. Immunol. 25, 441-449 KIESER, K. J. u. E. J. RUBIN (2014): How sisters grow apart: mycobacterial growth and division. Nat Rev Micro 12, 550-562

Page 152: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

148

KIM, S. Y., J. R. GOODMAN, M. PETROFSKY u. L. E. BERMUDEZ (1998): Mycobacterium avium infection of gut mucosa in mice associated with late inflammatory response and intestinal cell necrosis. J. Med. Microbiol. 47, 725-731 KLANG, A., C. STAFFLER, C. MASCHERBAUER, J. SPERGSER, B. C. RUTGEN, B. HINNEY, N. LUCKSCHANDER-ZELLER u. F. KUNZEL (2014): Mycobacterium avium subspecies hominissuis infection in a domestic European shorthair cat. Wien. Tierarztl. Monatsschr. 101, 74-78 KLANICOVA-ZALEWSKA, B. u. I. SLANA (2014): Presence and persistence of Mycobacterium avium and other nontuberculous mycobacteria in animal tissues and derived foods: A review. Meat Science 98, 835-841 KLANICOVA, B., I. SLANA, H. VONDRUSKOVA, M. KAEVSKA u. I. PAVLIK (2011): Real-time quantitative PCR detection of Mycobacterium avium subspecies in meat products. Journal of Food Protection® 74, 636-640 KÖHLER, H., A. SOSCHINKA, M. MEYER, A. KATHER, P. REINHOLD u. E. LIEBLER-TENORIO (2015): Characterization of a caprine model for the subclinical initial phase of Mycobacterium avium subsp. paratuberculosis infection. BMC Vet. Res. 11, 74 KOKOLUS, K. M., M. L. CAPITANO, C.-T. LEE, J. W.-L. ENG, J. D. WAIGHT, B. L. HYLANDER, S. SEXTON, C.-C. HONG, C. J. GORDON u. S. I. ABRAMS (2013): Baseline tumor growth and immune control in laboratory mice are significantly influenced by subthermoneutral housing temperature. Proceedings of the National Academy of Sciences 110, 20176-20181 KRIZ, P., P. JAHN, B. BEZDEKOVA, M. BLAHUTKOVA, V. MRLIK, I. SLANA u. I. PAVLIK (2010): Mycobacterium avium subsp. hominissuis infection in horses. Emerg. Infect. Dis. 16, 1328-1329 KRÜGER, C., H. KÖHLER u. E. M. LIEBLER-TENORIO (2015): Sequential Development of Lesions 3, 6, 9, and 12 Months After Experimental Infection of Goat Kids With Mycobacterium avium subsp paratuberculosis. Vet. Pathol. 52, 276-290 KRZYWINSKA, E. u. J. S. SCHOREY (2003): Characterization of genetic differences between Mycobacterium avium subsp. avium strains of diverse virulence with a focus on the glycopeptidolipid biosynthesis cluster. Vet. Microbiol. 91, 249-264

Page 153: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

149

KUNZE, Z. M., F. PORTAELS u. J. J. MCFADDEN (1992): Biologically distinct subtypes of Mycobacterium avium differ in possession of insertion sequence IS901. J. Clin. Microbiol. 30, 2366-2372 LAHIRI, A., J. KNEISEL, I. KLOSTER, E. KAMAL u. A. LEWIN (2014): Abundance of Mycobacterium avium ssp. hominissuis in soil and dust in Germany - implications for the infection route. Lett. Appl. Microbiol. 59, 65-70 LANZAS, C., P. AYSCUE, R. IVANEK u. Y. T. GROHN (2010): Model or meal? Farm animal populations as models for infectious diseases of humans. Nat Rev Micro 8, 139-148 LENAERTS, A., C. E. BARRY, 3RD u. V. DARTOIS (2015): Heterogeneity in tuberculosis pathology, microenvironments and therapeutic responses. Immunol. Rev. 264, 288-307 LENAERTS, A. J., D. HOFF, S. ALY, S. EHLERS, K. ANDRIES, L. CANTARERO, I. M. ORME u. R. J. BASARABA (2007): Location of persisting mycobacteria in a Guinea pig model of tuberculosis revealed by r207910. Antimicrob. Agents Chemother. 51, 3338-3345 LIEBANA, E., L. JOHNSON, J. GOUGH, P. DURR, K. JAHANS, R. CLIFTON-HADLEY, Y. SPENCER, R. G. HEWINSON u. S. H. DOWNS (2008): Pathology of naturally occurring bovine tuberculosis in England and Wales. The Veterinary Journal 176, 354-360 LIEBANA, E., S. MARSH, J. GOUGH, A. NUNEZ, H. M. VORDERMEIER, A. WHELAN, Y. SPENCER, R. CLIFTON-HARDLEY, G. HEWINSON u. L. JOHNSON (2007): Distribution and activation of T-lymphocyte subsets in tuberculous bovine lymph-node granulomas. Vet. Pathol. 44, 366-372 MANCUSO, J. D., G. H. MAZUREK, D. TRIBBLE, C. OLSEN, N. E. ARONSON, L. GEITER, D. GOODWIN u. L. W. KEEP (2012): Discordance among Commercially Available Diagnostics for Latent Tuberculosis Infection. Am. J. Respir. Crit. Care Med. 185, 427-434

Page 154: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

150

MARAIS, B., R. GIE, H. SCHAAF, A. HESSELING, C. OBIHARA, J. STARKE, D. ENARSON, P. DONALD u. N. BEYERS (2004): The natural history of childhood intra-thoracic tuberculosis: a critical review of literature from the pre-chemotherapy era [State of the Art]. The International Journal of Tuberculosis and Lung Disease 8, 392-402 MATLOVA, L., L. DVORSKA, W. Y. AYELE, M. BARTOS, T. AMEMORI u. I. PAVLIK (2005): Distribution of Mycobacterium avium complex isolates in tissue samples of pigs fed peat naturally contaminated with mycobacteria as a supplement. J. Clin. Microbiol. 43, 1261-1268 MATTY, M. A., F. J. ROCA, M. R. CRONAN u. D. M. TOBIN (2015): Adventures within the speckled band: heterogeneity, angiogenesis, and balanced inflammation in the tuberculous granuloma. Immunol. Rev. 264, 276-287 MENIN, Á., R. FLEITH, C. RECK, M. MARLOW, P. FERNANDES, C. PILATI u. A. BÁFICA (2013): Asymptomatic cattle naturally infected with Mycobacterium bovis present exacerbated tissue pathology and bacterial dissemination. PLoS ONE 8, e53884 MIJS, W., P. DE HAAS, R. ROSSAU, T. VAN DER LAAN, L. RIGOUTS, F. PORTAELS u. D. VAN SOOLINGEN (2002): Molecular evidence to support a proposal to reserve the designation Mycobacterium avium subsp avium for bird-type isolates and 'M-avium subsp hominissuis' for the human/porcine type of M-avium. Int. J. Syst. Evol. Microbiol. 52, 1505-1518 MÖBIUS, P., P. LENTZSCH, I. MOSER, L. NAUMANN, G. MARTIN u. H. KÖHLER (2006): Comparative macrorestriction and RFLP analysis of Mycobacterium avium subsp. avium and Mycobacterium avium subsp. hominissuis isolates from man, pig, and cattle. Vet. Microbiol. 117, 284-291 MOMOTANI, E., D. L. WHIPPLE, A. B. THIERMANN u. N. F. CHEVILLE (1988): Role of M cells and macrophages in the entrance of Mycobacterium paratuberculosis into domes of ileal Peyer's patches in calves. Vet. Pathol. 25, 131-137 MORAVKOVA, M., V. MRLIK, I. PARMOVA, P. KRIZ u. I. PAVLIK (2013): High incidence of Mycobacterium avium subspecies hominissuis infection in a zoo population of bongo antelopes (Tragelaphus eurycerus). J. Vet. Diagn. Invest. 25, 531-534

Page 155: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

151

MUKHERJEE, R. u. D. CHATTERJI (2012): Glycopeptidolipids: immuno-modulators in greasy mycobacterial cell envelope. IUBMB Life 64, 215-225 NIEBERLE, K. (1937): Über die Infektionswege der tuberkulösen Erkrankung bei jungen Kälbern. Arch. wiss. prakt. Tierhlk. 71, p. 323 NISHIMORI, K., M. EGUCHI, Y. NAKAOKA, Y. ONODERA, T. ITO u. K. TANAKA (1995): Distribution of IS901 in strains of Mycobacterium avium complex from swine by using IS901-detecting primers that discriminate between M. avium and Mycobacterium intracellulare. J. Clin. Microbiol. 33, 2102-2106 O'GARRA, A., P. S. REDFORD, F. W. MCNAB, C. I. BLOOM, R. J. WILKINSON u. M. P. BERRY (2013): The immune response in tuberculosis. Annu. Rev. Immunol. 31, 475-527 O’CONNELL, M. L., K. E. BIRKENKAMP, D. E. KLEINER, L. R. FOLIO, S. M. HOLLAND u. K. N. OLIVIER (2012): Lung manifestations in an autopsy-based series of pulmonary or disseminated nontuberculous mycobacterial disease. CHEST Journal 141, 1203-1209 OLSEN, I., T. B. JOHANSEN, H. BILLMAN-JACOBE, S. F. NILSEN u. B. DJONNE (2004): A novel IS element, ISMpa1, in Mycobacterium avium subsp. paratuberculosis. Vet. Microbiol. 98, 297-306 ORME, I. M. u. R. J. BASARABA (2014): The formation of the granuloma in tuberculosis infection. Semin. Immunol. 26, 601-609 ORME, I. M. u. D. J. ORDWAY (2014): Host Response to Nontuberculous Mycobacterial Infections of Current Clinical Importance. Infect. Immun. 82, 3516-3522 PALANISAMY, G. S., E. E. SMITH, C. A. SHANLEY, D. J. ORDWAY, I. M. ORME u. R. J. BASARABA (2008): Disseminated disease severity as a measure of virulence of Mycobacterium tuberculosis in the guinea pig model. Tuberculosis 88, 295-306

Page 156: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

152

PALMER, M. V., T. C. THACKER u. W. R. WATERS (2015): Analysis of Cytokine Gene Expression using a Novel Chromogenic In-situ Hybridization Method in Pulmonary Granulomas of Cattle Infected Experimentally by Aerosolized Mycobacterium bovis. J. Comp. Pathol. 153, 150-159 PALMER, M. V., W. R. WATERS u. T. C. THACKER (2007): Lesion development and immunohistochemical changes in granulomas from cattle experimentally infected with Mycobacterium bovis. Vet. Pathol. 44, 863-874 PESCIAROLI, M., J. ALVAREZ, M. B. BONIOTTI, M. CAGIOLA, V. DI MARCO, C. MARIANELLI, M. PACCIARINI u. P. PASQUALI (2014): Tuberculosis in domestic animal species. Res. Vet. Sci. 97 Suppl, S78-85 PETERS, W. u. J. D. ERNST (2003): Mechanisms of cell recruitment in the immune response to Mycobacterium tuberculosis. Microbes Infect. 5, 151-158 PETROFSKY, M. u. L. E. BERMUDEZ (2005): CD4+ T cells but Not CD8+ or gammadelta+ lymphocytes are required for host protection against Mycobacterium avium infection and dissemination through the intestinal route. Infect. Immun. 73, 2621-2627 PITZALIS, C., G. W. JONES, M. BOMBARDIERI u. S. A. JONES (2014): Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat. Rev. Immunol. 14, 447-462 RADOMSKI, N., V. C. THIBAULT, C. KAROUI, K. DE CRUZ, T. COCHARD, C. GUTIERREZ, P. SUPPLY, F. BIET u. M. L. BOSCHIROLI (2010): Determination of genotypic diversity of Mycobacterium avium subspecies from human and animal origins by mycobacterial interspersed repetitive-unit-variable-number tandem-repeat and IS1311 restriction fragment length polymorphism typing methods. J. Clin. Microbiol. 48, 1026-1034 RAMAKRISHNAN, L. (2012): Revisiting the role of the granuloma in tuberculosis. Nat. Rev. Immunol. 12, 352-366 RHOADES, E., A. FRANK u. I. ORME (1997): Progression of chronic pulmonary tuberculosis in mice aerogenically infected with virulent Mycobacterium tuberculosis. Tuber. Lung Dis. 78, 57-66

Page 157: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

153

RINDI, L. u. C. GARZELLI (2014): Genetic diversity and phylogeny of Mycobacterium avium. Infect. Genet. Evol. 21, 375-383 ROCCO, J. u. V. IRANI (2011): Mycobacterium avium and modulation of the host macrophage immune mechanisms [Review article]. The International Journal of Tuberculosis and Lung Disease 15, 447-452 RODRÍGUEZ, E., L. SÁNCHEZ, S. PÉREZ, L. HERRERA, M. JIMÉNEZ, S. SAMPER u. M. IGLESIAS (2009): Human tuberculosis due to Mycobacterium bovis and M. caprae in Spain, 2004–2007. The International Journal of Tuberculosis and Lung Disease 13, 1536-1541 ROSE, S. J. u. L. E. BERMUDEZ (2014): Mycobacterium avium Biofilm Attenuates Mononuclear Phagocyte Function by Triggering Hyperstimulation and Apoptosis during Early Infection. Infect. Immun. 82, 405-412 ROTH, R. I., R. L. OWEN, D. F. KEREN u. P. A. VOLBERDING (1985): Intestinal infection withMycobacterium avium in acquired immune deficiency syndrome (AIDS). Dig. Dis. Sci. 30, 497-504 SANCHEZ, J., L. TOMÁS, N. ORTEGA, A. J. BUENDÍA, L. DEL RIO, J. SALINAS, J. BEZOS, M. R. CARO u. J. A. NAVARRO (2011): Microscopical and Immunological Features of Tuberculoid Granulomata and Cavitary Pulmonary Tuberculosis in Naturally Infected Goats. J. Comp. Pathol. 145, 107-117 SANGARI, F. J., J. GOODMAN, M. PETROFSKY, P. KOLONOSKI u. L. E. BERMUDEZ (2001): Mycobacterium avium invades the intestinal mucosa primarily by interacting with enterocytes. Infect. Immun. 69, 1515-1520 SCHOREY, J. S. u. L. SWEET (2008): The mycobacterial glycopeptidolipids: structure, function, and their role in pathogenesis. Glycobiology 18, 832-841 SECOTT, T. E., T. L. LIN u. C. C. WU (2004): Mycobacterium avium subsp. paratuberculosis fibronectin attachment protein facilitates M-cell targeting and invasion through a fibronectin bridge with host integrins. Infect. Immun. 72, 3724-3732

Page 158: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

154

SEVILLA, I. A., E. MOLINA, N. ELGUEZABAL, V. PÉREZ, J. M. GARRIDO u. R. A. JUSTE (2015): Detection of Mycobacteria, Mycobacterium avium Subspecies, and Mycobacterium tuberculosis Complex by a Novel Tetraplex Real-Time PCR Assay. J. Clin. Microbiol. 53, 930-940 SHALER, C. R., C. N. HORVATH, M. JEYANATHAN u. Z. XING (2013): Within the Enemy’s Camp: contribution of the granuloma to the dissemination, persistence and transmission of Mycobacterium tuberculosis. Frontiers in Immunology 4, 30 SHARPE, A. E., C. P. BRADY, A. J. JOHNSON, W. BYRNE, K. KENNY u. E. COSTELLO (2010): Concurrent outbreak of tuberculosis and caseous lymphadenitis in a goat herd. Vet. Rec. 166, 591-592 SIGURÐARDÓTTIR, Ó. G., C. M. PRESS u. O. EVENSEN (2001): Uptake of Mycobacterium avium subsp. paratuberculosis through the distal small intestinal mucosa in goats: an ultrastructural study. Vet. Pathol. 38, 184-189 STIRLING, J., M. GRIFFITH, J. S. DOOLEY, C. E. GOLDSMITH, A. LOUGHREY, C. J. LOWERY, R. MCCLURG, K. MCCORRY, D. MCDOWELL, A. MCMAHON, B. C. MILLAR, J. RAO, P. J. ROONEY, W. J. SNELLING, M. MATSUDA u. J. E. MOORE (2008): Zoonoses associated with petting farms and open zoos. Vector Borne Zoonotic Dis. 8, 85-92 STOOP, E. J., W. BITTER u. A. M. VAN DER SAR (2012): Tubercle bacilli rely on a type VII army for pathogenicity. Trends Microbiol. 20, 477-484 SWEET, L. u. J. S. SCHOREY (2006): Glycopeptidolipids from Mycobacterium avium promote macrophage activation in a TLR2- and MyD88-dependent manner. J. Leukoc. Biol. 80, 415-423 THIBAULT, V. C., M. GRAYON, M. L. BOSCHIROLI, C. HUBBANS, P. OVERDUIN, K. STEVENSON, M. C. GUTIERREZ, P. SUPPLY u. F. BIET (2007): New variable-number tandem-repeat markers for typing Mycobacterium avium subsp. paratuberculosis and M. avium strains: comparison with IS900 and IS1245 restriction fragment length polymorphism typing. J. Clin. Microbiol. 45, 2404-2410

Page 159: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

155

TICHENOR, W. S., J. THURLOW, S. MCNULTY, B. A. BROWN-ELLIOTT, R. J. WALLACE, JR. u. J. O. FALKINHAM, 3RD (2012): Nontuberculous Mycobacteria in household plumbing as possible cause of chronic rhinosinusitis. Emerg. Infect. Dis. 18, 1612-1617 TIZARD, I. R. (2012): Veterinary immunology. Elsevier Health Sciences, TOBIN, D. M., F. J. ROCA, S. F. OH, R. MCFARLAND, T. W. VICKERY, J. P. RAY, D. C. KO, Y. ZOU, N. D. BANG, T. T. CHAU, J. C. VARY, T. R. HAWN, S. J. DUNSTAN, J. J. FARRAR, G. E. THWAITES, M. C. KING, C. N. SERHAN u. L. RAMAKRISHNAN (2012): Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections. Cell 148, 434-446 TORTOLI, E. (2012): Phylogeny of the genus Mycobacterium: Many doubts, few certainties. Infect. Genet. Evol. 12, 827-831 TRAN, Q. T. u. X. Y. HAN (2014): Subspecies identification and significance of 257 clinical strains of Mycobacterium avium. J. Clin. Microbiol. 52, 1201-1206 TURENNE, C. Y., R. WALLACE, JR. u. M. A. BEHR (2007): Mycobacterium avium in the postgenomic era. Clin. Microbiol. Rev. 20, 205-229 TURNER, O. C., R. J. BASARABA u. I. M. ORME (2003): Immunopathogenesis of pulmonary granulomas in the guinea pig after infection with Mycobacterium tuberculosis. Infect. Immun. 71, 864-871 ULRICHS, T. u. S. H. KAUFMANN (2006): New insights into the function of granulomas in human tuberculosis. J. Pathol. 208, 261-269 ULRICHS, T., G. A. KOSMIADI, S. JORG, L. PRADL, M. TITUKHINA, V. MISHENKO, N. GUSHINA u. S. H. KAUFMANN (2005): Differential organization of the local immune response in patients with active cavitary tuberculosis or with nonprogressive tuberculoma. J. Infect. Dis. 192, 89-97

Page 160: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

156

ULRICHS, T., G. A. KOSMIADI, V. TRUSOV, S. JORG, L. PRADL, M. TITUKHINA, V. MISHENKO, N. GUSHINA u. S. H. KAUFMANN (2004): Human tuberculous granulomas induce peripheral lymphoid follicle-like structures to orchestrate local host defence in the lung. J. Pathol. 204, 217-228 UZAL, F. A. u. J. G. SONGER (2008): Diagnosis of Clostridium perfringens intestinal infections in sheep and goats. J. Vet. Diagn. Invest. 20, 253-265 VAN RHIJN, I., J. GODFROID, A. MICHEL u. V. RUTTEN (2008): Bovine tuberculosis as a model for human tuberculosis: advantages over small animal models. Microbes Infect. 10, 711-715 VAN SOOLINGEN, D., P. W. HERMANS, P. E. DE HAAS, D. R. SOLL u. J. D. VAN EMBDEN (1991): Occurrence and stability of insertion sequences in Mycobacterium tuberculosis complex strains: evaluation of an insertion sequence-dependent DNA polymorphism as a tool in the epidemiology of tuberculosis. J. Clin. Microbiol. 29, 2578-2586 VELMURUGAN, K., B. CHEN, J. L. MILLER, S. AZOGUE, S. GURSES, T. HSU, M. GLICKMAN, W. R. JACOBS JR, S. A. PORCELLI u. V. BRIKEN (2007): Mycobacterium tuberculosis nuoG is a virulence gene that inhibits apoptosis of infected host cells. VIA, L. E., P. L. LIN, S. M. RAY, J. CARRILLO, S. S. ALLEN, S. Y. EUM, K. TAYLOR, E. KLEIN, U. MANJUNATHA u. J. GONZALES (2008): Tuberculous granulomas are hypoxic in guinea pigs, rabbits, and nonhuman primates. Infect. Immun. 76, 2333-2340 VILAPLANA, C. u. P.-J. CARDONA (2014): The lack of a big picture in tuberculosis: the clinical point of view, the problems of experimental modeling and immunomodulation. The factors we should consider when designing novel treatment strategies. Frontiers in microbiology 5, 55 VORDERMEIER, H. M., M. A. CHAMBERS, B. M. BUDDLE, J. M. POLLOCK u. R. G. HEWINSON (2006): Progress in the development of vaccines and diagnostic reagents to control tuberculosis in cattle. The Veterinary Journal 171, 229-244

Page 161: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

References

157

VORDERMEIER, H. M., M. A. CHAMBERS, P. J. COCKLE, A. O. WHELAN, J. SIMMONS u. R. G. HEWINSON (2002): Correlation of ESAT-6-specific gamma interferon production with pathology in cattle following Mycobacterium bovis BCG vaccination against experimental bovine tuberculosis. Infect. Immun. 70, 3026-3032 WANGOO, A., L. JOHNSON, J. GOUGH, R. ACKBAR, S. INGLUT, D. HICKS, Y. SPENCER, G. HEWINSON u. M. VORDERMEIER (2005): Advanced Granulomatous Lesions in Mycobacterium bovis-infected Cattle are Associated with Increased Expression of Type I Procollagen, γδ (WC1+) T Cells and CD 68+ Cells. J. Comp. Pathol. 133, 223-234 WATERS, W. R., M. F. MAGGIOLI, J. L. MCGILL, K. P. LYASHCHENKO u. M. V. PALMER (2014): Relevance of bovine tuberculosis research to the understanding of human disease: Historical perspectives, approaches, and immunologic mechanisms. Vet. Immunol. Immunopathol. 159, 113-132 WATERS, W. R., M. V. PALMER, B. M. BUDDLE u. H. M. VORDERMEIER (2012): Bovine tuberculosis vaccine research: Historical perspectives and recent advances. Vaccine 30, 2611-2622 WEISS, C. H. u. J. GLASSROTH (2012): Pulmonary disease caused by nontuberculous mycobacteria. Expert Rev Respir Med 6, 597-612; quiz 613 WOLF, A. J., L. DESVIGNES, B. LINAS, N. BANAIEE, T. TAMURA, K. TAKATSU u. J. D. ERNST (2008): Initiation of the adaptive immune response to Mycobacterium tuberculosis depends on antigen production in the local lymph node, not the lungs. The Journal of experimental medicine 205, 105-115 WOLINSKY, E. (1992): Mycobacterial diseases other than tuberculosis. Clin. Infect. Dis. 15, 1-12 YOUNG, D. (2009): Animal models of tuberculosis. Eur. J. Immunol. 39, 2011-2014

Page 162: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Annex

158

10 Annex

Neutral buffered Formalin according to Lillie (4%)

Mix 900 ml of distilled water with 100 ml 37% formalin (Merck KGaA, Darmstadt,

Germany), 4,0 g NaH2PO4 x H2O24(VWR International GmbH, Darmstadt, Germany)

and 16,4 g Na2HPO4 x 12H2O (VWR International GmbH, Darmstadt, Germany).

Coating of slides

For FFPE sections:

Gelatin:

Disolve 0.45 g of gelatin (Sigma-Aldrich Chemie GmbH, Steinheim, Germany) in 100

ml distilled water at 80°C, cool down and add five granules of thymol (Thüringer

Universitätsapotheke, Jena, Germany).

Chrome alum solution:

Disolve 160 mg of chrome alum (CrK(SO4)2 x 12 H20, VEB Laborchemie, Apolda,

GDR) in 4.0 ml of distilled water.

Working solution:

Mix 100 ml disolved gelatin and 3.85 ml of chrome alum solution.

For frozen sections:

Disolve 0.625g gelatin in 250 ml of distilled water at 80 °C and let cool down.

Add 0.063 g chrome alum and one to five granules of thymol.

Histochemical staining

Hemalum-eosin staining

Mayer´s hemalum:

Page 163: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Annex

159

Disolve 1.0 g of hematoxylin (Riedel-de Haen AG, Seelze-Hannover, Germany) in

1000 mlof distilled water, add 0.2 g sodium jodate (NAJO3), (Merck KGaA,

Darmstadt, Germany) and 50 g potassium alum (VEB Laborchemie, Apolda, GDR)

and disolve by shaking. Add 50 g of cloral hydrate (C2H3Cl3O2), (Carl Roth GmbH &

Co. KG, Karlsruhe, Germany) and 1.0 g of crystalized citric acid (VWR International

GmbH, Darmstadt, Germany) and filtrate.

1 % eosin solution:

Disolve 1.0 g of Eosin (Carl Roth GmbH & Co. KG, Karlsruhe, Germany) in 50%

distilled ethanol (mix absolute ethanol 1:2 with aqua bidest). Finally, add one drop of

glacial acetic acid (Merck KGaA, Darmstadt, Germany) immediately before use of

solution and filtrate.

Carbolic xylene:

Disolve 10 g of phenol (Merck KGaA, Darmstadt, Germany) in 100ml of xylene (Dr.

K. Hollborn & Söhne GmbH & Co. KG, Leipzig, Germany).

Azan stain

Azocarmine B solution:

Add 0.5 g of azocarmine B (unknown, GDR)to 100ml of distilled water, briefly cook

and filtrate.

Aniline alcohol:

Dilute 0.1 ml of aniline (Sigma-Aldrich Chemie GmbH, Steinheim, Germany) in 100ml

of 96% ethanol.

1% Acetic alcohol:

Dilute 1.0 ml glacial acetic acid in 100 ml of 96% ethanol.

Page 164: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Annex

160

Aniline blue/ orange G solution:

stock solution: Mix 0.5 g of aniline blue (Cheampol, Prag, Czechoslovakia), 2.0 g

Orange G (Feinchemie KG, Sebnitz, GDR), 100ml of distilled water and 8ml of 1%

acetic alcohol, heat, cool down and filtrate.

Working solution: dilute stock solution 1:2 with distilled water.

Immunohistochemistry

PBS buffer:

Stock solution (pH 6.8):

Disolve 40 g NaCl (Merck KGaA, Darmstadt, Germany) and 10 g of NaH2PO4 x 2H2O

(VWR International GmbH, Darmstadt, Germany)in 1000 ml of distilled water. Adjust

the solution with NaOH to pH 6.8.

Working solution (pH 7.1):

Mix 200 ml of stock solution with 800 ml of distilled water.

Tris-PBS-buffer:

Stock solution (pH 7.6):

Disolvein 60.57 g of Tris (Carl Roth GmbH & Co. KG, Karlsruhe, Germany)in 700 ml

of distilled water and adjust solution with 25 % hydrochloric acid (HCl) to pH 7.6 and

fill up with distilled water to 1000ml.

Working solution (pH 7.3):

Mix 100 ml of stock solution with 900 ml of PBS working solution (pH 7.1).

Acetate buffer (0.1M, pH 5.2)

0.1 N glacial acetic acid:

Dilute 2.87 ml glacial acetic acid in 500ml distilled water.

0.1 M sodium acetate:

Page 165: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Annex

161

Dilute 8.166 g sodium acetate (Merck KGaA, Darmstadt, Germany) in 100 ml of

distilled water.

Buffer solution:

Mix 210 ml of 0.1N glacial acetic acid with 790 ml of sodium acetate, adjust to pH

5.2.

Dulbecco´s PBS (DPBS)-buffer

Stock solution:

Disolve 40 g NaCl, 1.0 g KCl (Carl Roth GmbH & Co. KG, Karlsruhe, Germany), 1.0 g

KH2PO4 (Merck KGaA, Darmstadt, Germany) and 14.39 g of NaH2PO4 x 12 H20 in

1000 ml of distilled water.

Working solution (pH7.4):

Dilute 200 ml of DPBS stock solution in 800 ml of distilled water.

12,5% BSA in DPBS (BSA/DPBS)

Disolve 12.5 g BSA in DPBS working solution.

0.06& phenylhydrazine solution

Dilute 30 µl phenylhydrazine (Sigma-Aldrich Chemie GmbH, Steinheim, Germany) in

50 ml of PBS working solution.

AEC-solution

Disolve 8.0 mg of AEC (Sigma-Aldrich Chemie GmbH, Steinheim, Germany) in 1.0

ml N,N-Dimethylformamid (Sigma-Aldrich Chemie GmbH, Steinheim, Germany). Add

1.0 ml solved AEC to 14 ml of acetate buffer and add 150 µl of 3% H2O2, filtrate and

use immediately.

3,3′-diaminobenzidine-tetrahydrochloride-dihydrate (DAB) solution

Disolve 24 mg of DAB (Sigma-Aldrich Chemie GmbH, Steinheim, Germany) in 40 ml

of PBS working solution. Add 400 µl of 3% H2O2, filtrate and use immediately.

Page 166: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Annex

162

0.01% osmic acid solution

Dilute1ml of 1% osmiumtetraoxide (OsO4, Carl Roth GmbH & Co. KG, Karlsruhe,

Germany) in 100 ml of distilled water.

2% methyl-green solution

Disolve 2.0 g methyl green (Sigma-Aldrich Chemie GmbH, Steinheim, Germany) in

100 ml of distilled water and filtrate twice, can be used repeatedly.

Page 167: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

Acknowledgements

163

11 Acknowledgements

Finally, I want to sincerely thank:

Prof. Dr. Elisabeth M. Liebler-Tenorio and Dr. Heike Köhler for providing the topic,

their invaluable support during the animal trial, the experiments, the publications, and

for providing a trustful environment.

Sabine Lied, Lisa Wirker and Lisa Wolf for their outstanding technical expertise and

for many good conversations and support during this thesis.

The staff of the animal facility of the FLI, Jena, for support during the animal trial.

Wolfram Maginot for his friendship, his fatherly advice and for his support during the

preparation of any kind of photographical data and layout decisions.

Petra Möbius for many good conversations and her support in a joint publication.

Prof. Dr. Dr. Petra Reinhold and Sina Fischer for good scientific cooperation and

constructive conversations during all working group meetings.

Maria-Christina Haase for providing enormous quantities of literature and text books

and all my colleagues for many good conversations and support during this thesis.

My brother, my parents, my grandparents, my parents in law and my friends for all

their support during the preparation of thesis.

Romy for her unlimited love, understanding and support without this thesis would

never have been completed.

Page 168: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2
Page 169: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2
Page 170: Mycobacterium avium subsp. hominissuis · LP lamina propria mucosae LPS lipopolysaccharide m meter M. ... TB tuberculosis TcR T-cell-receptor TGF transforming growth factor Th1/Th2

ISBN 978-3-86345-288-9

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

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

Jan

Sch

inkö

the H

an

no

ver

2015

Experimental oral infection of goats with

Mycobacterium avium subsp. hominissuis:

Pathomorphological characterization of lesions in

different courses of disease with special focus on

cellular composition of granulomas

Jan Schinköthe

Hannover 2015


Recommended