114
Research Collection Doctoral Thesis Approach to the gene controlling the porcine receptor for Escherichia coli with fimbriae F4ab/F4ac and inheritance of the receptor for F4ad Author(s): Rampoldi, Antonio Publication Date: 2013 Permanent Link: https://doi.org/10.3929/ethz-a-009907149 Rights / License: In Copyright - Non-Commercial Use Permitted This page was generated automatically upon download from the ETH Zurich Research Collection . For more information please consult the Terms of use . ETH Library

Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

Research Collection

Doctoral Thesis

Approach to the gene controlling the porcine receptor forEscherichia coli with fimbriae F4ab/F4ac and inheritance of thereceptor for F4ad

Author(s): Rampoldi, Antonio

Publication Date: 2013

Permanent Link: https://doi.org/10.3929/ethz-a-009907149

Rights / License: In Copyright - Non-Commercial Use Permitted

This page was generated automatically upon download from the ETH Zurich Research Collection. For moreinformation please consult the Terms of use.

ETH Library

Page 2: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 1 -

DISS. ETH No. 21052

Approach to the gene controlling the porcine receptor for Escherichia coli with

fimbriae F4ab/F4ac and inheritance of the receptor for F4ad

A dissertation submitted to

ETH ZURICH

for the degree of

Doctor of Sciences

presented by

Antonio Rampoldi

Master’s Degree in Veterinary Biotechnology, University of Milan

Born March 26, 1983

Citizen of Italy

accepted on the recommendation of

Prof. Dr. P. Vögeli, examiner

Prof. Dr. M. Kreuzer, co-examiner

PD Dr. S. Neuenschwander, co-examiner

Prof. Dr. H.U. Bertschinger, co-examiner

2013

Page 3: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 2 -

"I like pigs. Dogs look up to us. Cats look down on us.

Pigs treat us as equals."

Sir Winston Churchill, UK Prime Minster (1874-1965)

Page 4: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 3 -

ACKNOWLEDGEMENTS

I would like to thank Prof. Dr. Peter Vögeli, former head of our group at the Institut für

Agrarwissenschaften (IAS) ETH Zurich, for giving me the opportunity to perform my doctorate in his

group and for supervising my work.

Sincere thanks go to PD Dr. Stefan Neuenschwander for his advice, supervision, solving

problems in the lab, and helping revise the publication.

I am very grateful to Prof. Dr. Hans Ulrich Bertschinger for his huge amount of work in

phenotyping of the intestinal samples and reviewing my thesis.

I thank Dr. Esther Bürgi and her staff from the Faculty of Veterinary Medicine, Vetsuisse

Faculty, University of Zurich, for taking care of the pigs, managing the piggery, and organizing the

slaughtering.

I thank also Andreas Hofer, Henning Luther and all the people from SUISAG for their interest in

financing our project on ETEC F4 susceptibility in pigs, and for providing the animals for our research.

A sincere thank you to Prof. Dr. Gaudenz Dolf of the Institute of Genetics, University of Bern

for the support in statistics on F4ad adhesion.

I would like to thank also Dr. Claus B. Jørgensen, Mette J. Jacobsen, and their group at the

University of Copenhagen for their collaboration in the E. coli F4 project, for sharing the results and data

of their research on pig susceptibility to ETEC F4 with our group.

I am also grateful for the help I received from the people working at the Genetic Diversity Centre

(GDC), ETH Zurich, and the Functional Genomics Center Zurich (FGCZ), a joint state-of-the-art facility

of the ETH Zurich and the University of Zurich.

I want to thank all the people who are or were part of our group:

• Gerda Bärtschi and Elisabeth Wenk for taking care of the house,

• Anna Bratus, Benita Pineroli, Bruno Dietrich and Monika Haubitz for their support and

interesting conversations,

• David Joller for the technical and personal assistance,

• Dr. Michael Goe for his English correction,

• Dr Markus Schneeberger,

Page 5: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 4 -

• Dr Johannes Kaiser,

• Dr Claude Schelling and Dr Aldona Pienkowska-Schelling,

• The student apprentices, especially Martin Stüssi, to whom I wish good luck on his future career.

I want also to thank my family, especially my parents and brother, and also my friends, both the

ones in Italy and the new ones I made in Zurich, for always encouraging me to keep going forward.

This project was financed by the Swiss National Science Foundation (no. 3100A0-120255/1), the

ETH Zurich and the SUISAG.

Page 6: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 5 -

CONTENTS

ACKNOWLEDGEMENTS 3

SUMMARY 8

SOMMARIO 10

1. INTRODUCTION 12

1.1 Diarrhoea 12

1.2 Escherichia coli 12

1.3 ETEC 14

1.3.1 Enterotoxins 14

1.3.2 Fimbriae 15

1.3.2.1 Fimbriae F4 15

1.3.2.2 Prevalence of ETEC F4 16

1.4 Determination of F4 receptor phenotypes 16

1.4.1 Alternative methods 17

1.5 ETEC F4 RECEPTORS 18

1.5.1 ETEC F4 receptor phenotypes in pigs 18

1.5.2 ETEC F4ab and ETEC F4ac receptor 18

1.5.3 ETEC F4ad receptors 19

1.5.4 ETEC F4ac susceptibility among breeds 19

1.6 Methods for preventing F4 diarrhoea 20

1.6.1 Antibiotics 20

1.6.2 Homeopathy 20

1.6.3 Immune protection by colostrum and milk 21

1.6.4 Vaccine 22

1.7 Genetic mapping of F4bcR 23

1.8 Candidate genes for F4bcR 24

1.8.1 Positional candidate gene 25

1.8.1.1 SLC12A8 25

1.8.2 Positional and functional candidate genes 25

1.8.2.1 HEG1 25

1.8.2.2 MUC13 26

1.8.2.3 ITGB5 26

1.9 Objectives 27

2. MATERIALS AND METHODS 28

2.1 Pigs 28

2.2 Determination of the ETEC F4 receptor phenotype 29

2.2.1 Preparation of ETEC F4 strains 29

Page 7: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 6 -

2.2.2 Sampling of intestinal tissues 29

2.2.3 Preparation of enterocytes 30

2.2.4 Microscopic adhesion test 30

2.2.5 Positive and negative control of bacterial adhesion 31

2.3 DNA methods 32

2.3.1 DNA extraction from blood 32

2.3.2 Polymerase chain reaction 33

2.3.2.1 Primer design 33

2.3.2.2 Standard PCR 33

2.3.2.3 Long-range PCR 37

2.3.2.4 PCR for pyrosequencing 39

2.3.3 Agarose gel electrophoresis 40

2.3.4 Genescan analysis 40

2.3.5 Pyrosequencing 41

2.3.6 SNP chip 42

2.3.7 PCR restriction 43

2.3.8 High throughput sequencing 45

2.3.8.1 Illumina HiSeq 2000 45

2.3.8.2 PacBio RS 46

2.3.9 PCR purification 47

2.4 RNA methods 48

2.4.1 RNA extraction 48

2.4.2 RNA quantification 48

2.4.3 Reverse Transcription PCR 49

2.4.4 PCR and sequencing 50

2.4.5 High throughput sequencing 50

2.5 Computational methods 51

2.5.1 Linkage analysis 51

2.5.2 Statistics of F4ad adhesion 51

2.5.3 In silico mapping 52

3. RESULTS 53

3.1 Exclusion of gene MUC4 as locus for F4bcR 53

3.2 Exclusion of interval ZDHHC19-LMLN as locus for F4bcR 56

3.3 SNPs chip results 59

3.4 Exclusion of interval LMLN-ZNF148 as locus for F4bcR 61

3.5 Exclusion of interval SLC12A8-KVL1293 as locus for F4bcR 63

3.6 Partial exclusion of MUC13 as locus for F4bcR 65

3.7 Sequencing and mRNA expression of candidate genes 67

3.7.1 SLC12A8 68

Page 8: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 7 -

3.7.2 HEG1 68

3.7.3 MUC13 69

3.7.4 ITGB5 70

3.8 Sequencing of intergenic regions 70

3.8.1 Interval of HEG1-MUC13 70

3.8.1 Interval of MUC13-ITGB5 71

3.9 Validation of alternative markers for ETEC F4ab/F4ac susceptibility 73

3.10 F4ad susceptibility 75

3.10.1 Two receptors for E. coli F4ad 75

3.10.2 Inheritance of phenotype 77

3.10.3 Statistical pedigree analysis 79

4. DISCUSSION 83

4.1 F4bcR mapping on SSC13 83

4.2 Exclusion of genes SLC12A8 and HEG1 83

4.3 Exclusion of genes ITGB5 84

4.3 MUC13 84

4.5 F4adR inheritance 85

4.6 Conclusions and perspectives 86

REFERENCES 87

ABBREVIATIONS 102

List of figures 103

List of tables 104

Appendix 105

Media and solutions 105

Chemicals 106

Restriction enzymes 107

Labware 107

SNPs nomenclature 109

CURRICULUM VITAE 111

Page 9: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 8 -

SUMMARY

The ability to colonise the intestine is a common feature of both pathogenic and non-pathogenic

bacteria. Enterotoxigenic E. coli (ETEC) is the major cause of diarrhoea and death among piglets. The

bacteria adhere to specific receptors on the brush borders of enterocytes by adhesive fimbriae, and they

subsequently produce toxins that stimulate diarrhoea. There are no more than five fimbrial types

described worldwide in vivo, among which F4 is the most prevalent in the world. The F4 fimbrial type

has three antigenic variants—F4ab, F4ac and F4ad—that vary in their receptor specificities.

In pigs, resistant or susceptible phenotypes for fimbriae F4ac are inherited as monogenetic traits,

the susceptible allele being dominant over the resistant one. The receptor for F4ac binds F4ab as well.

Genome scans with microsatellites have localised the ETEC F4ab/F4ac receptor gene (F4bcR) to a region

of pig chromosome 13 (SSC13), SSC13q41-q44. Mucin 4 (MUC4) and mucin 13 (MUC13) genes have

been mapped in SSC13q41-q44, and they co-segregate mostly with the F4bcR alleles. SNPs derived from

these two genes are being used in breeding programs to reduce the frequency of the susceptible allele for

ETEC F4ab/F4ac in the pig population.

In this study, selected pigs from the Swiss Performing Station (SPS) in Sempach and from the

university experimental herd (UEH) at the University of Zurich were found to be recombinant in the

MUC4-F4bcR or F4bcR-MUC13 intervals.

A three-generation study was performed on 32 selected pigs from UEH, recombinant in the

MUC4-F4bcR interval, with the use of the Porcine SNP60 DNA BeadChip. DNA of a pig recombinant in

the F4bcR-MUC13 interval was sequenced with high throughput techniques, using HiSeq 2000 and

PacBio RS platforms.

RNA samples of pigs resistant and susceptible to ETEC F4ab/F4ac were sequenced with a

SOLiD platform. Analyses were performed to investigate the expression and possible mRNA variation of

candidate genes’ HEG homolog 1 (HEG1), solute carrier family 12 member 8 (SLC12A8), MUC13, and

integrin β-5 (ITGB5) in the region SSC13q41-q44 in the small intestine.

Markers were tested in both susceptible and resistant pigs, looking for a linkage disequilibrium

(LD) with F4bcR. GeneScan analyses were performed on microsatellites. PCR-RFLP and pyrosequencing

were performed on SNPs with possible LD with F4bcR.

Page 10: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 9 -

The data obtained in this study have led to a further narrowing down of the locus of F4bcR to

exon 2 of MUC13 and to the discovery of several markers with high LD with F4bcR, to be used in

breeding programs: ALGA0072075, KVL1293, ALGA0106330, MUC13-226, and MUC13-813.

The inheritance of the receptor or receptors for F4ad instead is not well understood. Aside from

the fully resistant and susceptible adhesion phenotypes, such as the ones seen in F4ac and F4ab, some of

the UEH pigs showed a third phenotype in the adhesion tests, in which only some of the enterocytes

examined were without bacteria. In this weak susceptible phenotype, the enterocytes seem to become

more susceptible to F4ad the closer they get to the ileocaecal valve.

Analyses revealed that there is more than one receptor for ETEC F4ad fimbriae on the surface of

the enterocytes—a strong receptor, responsible for full adhesion phenotype (E1), and a weak receptor,

responsible for the low adhesion phenotype (E2).

Adhesion tests were performed on 489 pigs from UEH to determine the receptor phenotype for

ETEC F4ad. Statistical analyses were performed with Pedigree Analysis Package v. 4.0 software to

evaluate possible models of inheritance for ETEC F4ad receptor.

Results indicate that the E1 phenotype might be encoded by two complementary or epistatic

genes, while the E2 phenotype is inherited as a dominant monogenetic trait.

Page 11: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 10 -

SOMMARIO

L’abilità di colonizzare l’intestino è una caratteristica comune di batteri patogeni e non-patogeni.

Enterotoxigenic E. coli (ETEC) è la maggior causa di diarrea e morte fra i lattonzoli. I batteri

aderiscono a specifici recettori sull’orletto a spazzola degli enterociti tramite fimbrie adesive, e

successivamente producono tossine che stimolano la diarrea. Esitono non più di cinque tipi di fimbrie nel

mondo, tra cui F4 è la più prevalente. La fimbria F4 possiede tre varianti antigeniche—F4ab, F4ac e

F4ad—che differiscono nella loro specificità verso i recettori.

Nei maiali, i fenotipi di resistenza o suscettibilità alle fimbrie F4ac sono ereditati come un

carattere monogenetico, l’allele per la suscettibilità è dominante verso quello per la resistenza. Il recettore

per le fimbrie F4ac lega anche le F4ab. Genome scan di microsatelliti hanno localizzato il gene recettore

per ETEC F4ab/F4ac (F4bcR) in una regione del cromosoma 13 del maiale (SSC13), SSC13q41-q44. I

geni mucin 4 (MUC4) e mucin 13 (MUC13) sono stati mappati in SSC13q41-q44, e co-segregano

generalmente con gli alleli di F4bcR. SNP derivati da questi due geni sono utilizzati in programmi di

riproduzione per ridurre la frequenza dell’allele per la suscettibilità all’ETEC F4ab/F4ac nella

popolazione suina.

In questo studio, maiali selezionati dalla Swiss Performing Station (SPS) in Sempach e da una

mandria sperimentale (UEH) dell’Università di Zurigo sono risultati ricombinanti negli intervalli MUC4-

F4bcR o F4bcR-MUC13.

Uno studio su tre generazioni è stato fatto con 32 maiali selezionati dalla UEH, ricombinanti

nell’intervallo MUC4-F4bcR, tramite l’uso del Porcine SNP60 DNA BeadChip. Il DNA di un maiale

ricombinante nell’intervallo F4bcR-MUC13 è stato sequenziato con tecniche high throughput, usando le

piattaforme HiSeq 2000 e PacBio RS.

Campioni di RNA da maiali resistenti e suscettibili all’ETEC F4ab/F4ac sono stati sequenziati

usando la piattaforma SOLiD. Le analisi sono state condotte per investigare l’espressione dell’mRNA e

possibili sue variazioni nell’intestino tenue sui geni candidati HEG homolog 1 (HEG1), solute carrier

family 12 member 8 (SLC12A8), MUC13, ed integrin β-5 (ITGB5) mappati nella regione SSC13q41-q44.

Page 12: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 11 -

I marker sono stati testati sia in maiali suscettibili che resistenti, cercando un disequilibrium da

linkage (LD) con F4bcR. GeneScan sono state eseguite su microsatelliti. PCR-RFLP e pyrosequencing

sono state eseguite su SNP con un possible LD con F4bcR.

I dati ottenuti in questo studio hanno ristretto il locus di F4bcR nell’esone 2 di MUC13 e portato

alla scoperta di diversi marker con un alto LD con F4bcR, da poter essere usati nei programmi di

riproduzione: ALGA0072075, KVL1293, ALGA0106330, MUC13-226, e MUC13-813.

L’eredità del recettore o dei recettori per F4ad invece non è ancora compresa. A parte i fenotipi

completamente resistenti o suscettibili all’adesione, come quelli osservati in F4ac e F4ab, alcuni maiali

dell’UEH mostravano un terzo fenotipo nei test di adesione, in cui solo alcuni degli enterociti esaminati

erano senza batteri. In questo fenotipo debolmente suscettibile, gli enterociti sembrano diventare più

suscettibili a F4ad più vicino sono alla valvola ileocecale.

Le analisi hanno rilevato che vi è più di un recettore per le fimbrie ETEC F4ad sulla superficie

degli enterociti—un recettore forte, responsabile per il fenotipo con una completa adesione (E1), e un

recettore debole, responsabile per il fenotipo con una bassa adesione (E2).

Test di adesione sono stati effettuati su 489 maiali dell’UEH per determinare il fenotipo del

recettore per ETEC F4ad. Le analisi statistiche sono state effettuate con il software Pedigree Analysis

Package v. 4.0 per valutare possibili modelli di ereditarietà per il recettore ETEC F4ad.

I risultati indicano che il fenotipo E1 può essere codificato da due geni complementari o

epistatici, mentre il fenotipo E2 è ereditato come un tratto dominante monogenetico.

Page 13: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 12 -

1. INTRODUCTION

1.1 Diarrhoea

The human body is composed of almost 100 trillion cells, and carries about 10 times as many

bacteria in the intestines (Björkstén et al., 2001; Steinhoff, 2005). At least 400 different bacterial species

are present in the gastrointestinal tract (Dunn, 1990; Moore & Holdeman, 1974).

The flora of the proximal small intestine differs significantly from that of the terminal ileum and

colon (Huijsdens et al., 2002). The most frequently identified anaerobic bacteria are Bacteroides spp.,

Bifidobacterium spp., Eubacterium spp., Peptostreptococcus spp., and Fusobacterium spp. (Simon &

Gorbach, 1986). These bacteria have a mostly a symbiotic relationship (Sears, 2005), contributing to the

host’s health through biotin, vitamin K, and hormones production (Guarner & Malagelada, 2003). In

certain conditions, pathogenic bacteria are able to replace the normal flora, causing diseases such as

diarrhoea.

Diarrhoea is a common problem for children in developing countries, for travellers (Clarke,

2001; Qadri et al., 2005), and in animal production. Pigs are susceptible to diarrhoea mainly in the first

five days after birth (neonatal period) and at four weeks (post-weaning period). The mortality rate of

piglets is 10%, and can be up to 25% if not treated (Kjaersgard et al., 2002; Li et al., 2007). The piglets

that survive grow more slowly and have poorer performance (Fairbrother & Gyles, 2006).

During the neonatal period, diarrhoea is associated with Escherichia coli, Clostridium

perfringens, or Isospora suis. In weaning piglets, diarrhoea is associated with Salmonella, Lawsonia

intracellularis, and Escherichia coli. Several viruses (e.g. rotavirus, calicivirus, astrovirus and

adenovirus) and parasites (nematodes, protozoa) can also cause water flow in the intestine. In addition,

diarrhoea can have other, non-related causes (e.g. digestive tract surgery, medications and malabsorption).

1.2 Escherichia coli

Escherichia coli (E. coli), a gram-negative bacterium from the Enterobacteriaceae family found

in the lower intestine, represent 0.1% of gut flora (Eckburg et al., 2005). Most E. coli strains are

Page 14: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 13 -

symbiotic to the host, producing vitamin K2 (Bentley & Meganathan, 1982) and preventing pathogenic

strains from colonising the intestine (Hudault et al., 2001; Reid et al., 2001).

The principal transmission means of pathogenic E. coli are faecal-oral. Pathogenic E. coli cause

severe infections and are occasionally responsible for food product recalls (Vogt & Dippold, 2005), as

occurred recently in Germany with a new strain resistant to antibiotics (Turner, 2011). Pathogenic E. coli

are responsible for 50% of all diarrhoea cases in post-weaning pigs (Gyles, 1994).

The pathogenic E. coli that causes diarrhoea is commonly divided into seven different

categories: enterotoxigenic E. coli (ETEC), enteropathogenic E. coli (EPEC), enterohaemorrhagic E. coli

(EHEC), vero cytotoxin-producing E. coli (VTEC), enteroaggregative E. coli (EAEC), enteroinvasive E.

coli (EIEC), and diffusely adherent E. coli (DAEC) (Nataro & Kaper, 1998). E. coli possess fimbrial

antigens that allow the bacteria to adhere to the enterocytes and colonise the intestine (MacKinnon, 1998)

(Figure 1.1).

Pigs are mostly affected by the ETEC, VTEC and EPEC strains. The ETEC strains contribute to

the majority of diarrhoea cases, while EPEC only contribute to 6% (Fairbrother, 1999; Markwalder,

2001).

Figure 1.1: E. coli bacterium with fimbriae (Gross, 2006)

Page 15: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 14 -

1.3 ETEC

ETEC carries adhesive fimbriae that attach to receptors on the brush border of the enterocytes

and produces enterotoxins that cause diarrhoea (Figure 1.2).

Figure 1.2: Small intestinal brush border with strong ETEC F4 adhesion (Bertschinger et al., 1972)

1.3.1 Enterotoxins

Enterotoxins produced by ETEC are divided into heat-labile enterotoxin (LT) and heat-stable

enterotoxin (ST). Enteroaggregative Escherichia coli heat-stable enterotoxin 1 (EAST1) produced by

EAEC is also associated with ETEC (Toledo et al., 2012; Zhao et al., 2009).

Heat-labile enterotoxins (LT-I and LT-II) are inactivated at high temperatures (Wagner et al.,

2004; Glenn et al., 2007). Subtype LT-I is found in animals and humans, and subtype LT-II is found only

in animals. The LT mechanism of action is similar to that of the cholera toxin (Brown & Hardwidge,

2007). LT raise cyclic AMP levels, increasing the secretion of chloride (Cl-) and the concentration of

calcium (Ca2+) and sodium (Na+) ions in the cells. The ion imbalance stimulates the flow of water into the

lumen, causing diarrhoea (Moon, 1978). Heat-stable enterotoxins (ST-a, ST-b, and EAST1) are resistant

to hydrolysis by gastric and jejunal enzymes and remain active at 100°C for 30 minutes (Kapitany et al.,

1979). EAST1 and ST raise cyclic GMP levels, which stimulate ion secretion, causing diarrhoea (Hughes

et al., 1978). Only the ETEC strains producing LT, EAST1, and ST-a are sufficiently pathogenic to cause

diarrhoea (Berberov et al., 2004; Zhang et al., 2006).

Page 16: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 15 -

1.3.2 Fimbriae

ETEC colonises the intestine by adhering to the enterocytes with its fimbriae. Only five fimbrial

types, F4 (formerly K88), F5 (K99), F6 (987P), F18 (F107), and F41, occur worldwide in pigs. Adhesin

involved in diffuse adherence (AIDA), an adhesin produced by DAEC, is also associated with ETEC

(Ngeleka et al., 2003; Zhao et al., 2009).

Fimbriae F6, F18, and F41 have a carbohydrate-binding protein site at their extremity that acts as

a binding site to the intestinal receptors (Moon, 1997). F4 and F5 have binding sites along their entire

length. ETEC expressing F5, F6, and F41 fimbriae are found mainly in the neonatal period, and ETEC

F18 is found mostly in the post-weaning period (Fairbrother et al., 2005; Nagy & Fekete, 2005). ETEC

expressing F4 fimbriae (ETEC F4) are responsible for diarrhoea in both periods. ETEC F4 and ETEC F18

are the most prominent worldwide. Genes controlling F4 and F18 fimbrial types were identified in 92.7%

of post-weaning ETEC diarrhoea cases (Frydendahl, 2002).

Pigs can be either susceptible or resistant to ETEC fimbriae adhesion, due to mutations in the

receptor proteins on the brush border (Sweeney, 1968). The mutation that makes pigs susceptible to

ETEC F18 is known (Meijerink et al., 1997; Meijerink et al., 2000). Elimination of the susceptible allele

for ETEC F18 from the porcine population is currently carried out in Switzerland and in other countries

(Luther et al., 2009).

1.3.2.1 Fimbriae F4

The F4 fimbriae are long, polymeric appendages composed of several hundred identical major

FaeG subunits (27.5 kDA each) with some minor subunits interspersed through the structure. One

bacterium can contain 100 to 300 fimbriae.

The F4 fimbriae are divided into three antigenic variants: F4ab, F4ac, and F4ad (previously

known as K88ab, K88ac, and K88ad). The variants share a common antigen (a) and express a specific

antigen (b, c, or d). The variants are distinguished by amino-acid substitutions in the major adhesive

subunit FaeG (Van Den Broeck et al., 2000; Guinée & Jansen, 1979) (Figure 1.3).

Page 17: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 16 -

Figure 1.3: Gene cluster encoding F4 fimbriae (Van der Broeck et al., 2000)

1.3.2.2 Prevalence of ETEC F4

Differences in fimbrial prevalence have been observed among neonatal, suckling, and weaning

piglets. ETEC F4 is the most important cause of diarrhoea in newborn and weaned piglets worldwide

(Frydendahl, 2002). In a report on 115 Swiss suckling piglets with diarrhoea, 50.5% of the samples were

positive for ETEC F4 (Sarrazin et al., 2000). In Denmark, ETEC F4 was present in 41% of all isolates

from 141 piglets with diarrhoea (Ojeniyi et al., 1994). In Hungary, ETEC F4 was present in 60% of all

isolates from 88 weaned pigs with diarrhoea (Nagy et al., 1990). In South Korea, 60% of 191 isolates

from piglets with diarrhoea were positive for genes of ETEC F4 (Kim et al., 2010). In North America,

65% of 175 piglets with diarrhoea were positive for genes of ETEC F4 (Zhang et al., 2007). In

Zimbabwe, of 67 neonatal piglets with diarrhoea, 28.4% were positive for genes of ETEC F4 (Madoroba

et al., 2009).

Recent vaccination programs for ETEC F4 and breeding selection may have changed the

fimbriae prevalence in ETEC, increasing the presence of other fimbrial types. In a report on 341 Saxonian

isolates from weaning piglets with diarrhoea, 15% of the samples were positive for F4 fimbriae (Wittig et

al., 1995). In Mexico, 3% of 935 isolates from suckling and weaned piglets with diarrhoea were positive

for ETEC F4 fimbriae (Toledo et al., 2012). In China, 9.8% of 215 isolates from pigs with post-weaning

diarrhoea were positive for ETEC F4 fimbriae (Chen et al., 2004a).

1.4 Determination of F4 receptor phenotypes

Three adhesion test methods have been reported for the determination of F4 phenotypes: 1)

screening for bacteria adhesion on brush border membrane vesicles prepared from enterocytes (Baker et

al., 1997; Sellwood et al., 1975); 2) screening of entire isolated enterocytes (Edfors-Lilja et al., 1986;

Page 18: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 17 -

Rapacz & Hasler-Rapacz, 1986); and 3) screening of intact intestinal villi (Cox & Houvenaghel, 1993;

Rasschaert et al., 2007). Bacterial adhesion to the brush border of the enterocytes was detected in all three

methods by light microscopy or phase contrast microscopy.

The threshold used in phenotyping for susceptibility to F4 fimbriae can affect the results of the

studies as well. Baker et al. (1997) prepared brush border vesicles, and specimens in which at least 10%

of 20 selected brush borders bound more than two bacteria were considered adhesive. Li et al. (2007)

used intact epithelial cells, and a cell was considered adhesive when more than five bacteria adhered to

the brush border and four degrees of adhesion strength were distinguished. Engel (1998) and Engel et al.

(1998) prepared epithelial cells, and pigs yielding not more than two of 10─60 cells binding more than

two bacteria were regarded as resistant.

An ELISA method can be used to determine the susceptibility or resistance to ETEC F4. The F4

fimbriae are bound to microtiter plates and then exposed to brush border vesicles, and adhesion is

revealed by antibodies against the brush borders (Chandler et al., 1994). This method results in 95%

correlation with the classical adhesion tests.

The adhesion test is an invasive method, usually conducted on dead piglets. Biopsies performed

on living pigs are time-consuming, expensive, and stressful for the pigs. As such, it is difficult to

incorporate these tests into breeding programs to select pigs resistant to ETEC expressing F4 fimbriae.

1.4.1 Alternative methods

Atroshi et al. (1983) showed a correlation between sow milk and susceptibility to ETEC F4.

Milk contains large quantities of fat globule membrane that may express receptors similar to those present

on the brush borders of intestinal cells. Alternatively, ETEC F4 may also bind to the immunoglobulins

(IgA) present on the globule membrane.

Valpotic et al. (1992) performed an enzyme immunoassay on brush borders collected from pig

faeces. The sensitivity of the assay was low in adult pigs, compared to neonatal and weaned piglets. The

faecal samples from adult pigs contained less receptor material than necessary for comparable

phenotyping.

Page 19: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 18 -

1.5 ETEC F4 RECEPTORS

1.5.1 ETEC F4 receptor phenotypes in pigs

Bijlsma et al. (1982) identified five ETEC F4 adhesion patterns in pigs, designated A-E. In

phenotype A, the three F4 fimbrial variants bind to the brush borders, and in phenotype E, none of the

variants bind (Table 1.1). Subsequently, new phenotypes were reported: phenotype F (Baker et al., 1997)

and phenotypes G and H, observed mainly in eastern breeds (Yan et al., 2009).

In phenotypes C and F, ETEC shows weak ETEC F4ab adhesion (Python et al., 2005). The weak

adhesion seen in the C and F phenotypes could be caused by an epistatic effect of some modifier genes,

by a case of RNA interference, or by an artefact. Similar conclusions regarding ETEC F4ac may apply in

phenotypes G and H.

Table 1.1: Phenotypes observed in pigs according to the binding of ETEC F4 variants A through H;

bacterial adhesion is marked with ● (Jacobsen, 2011-modified version).

Phenotypes Fimbrial variants

F4ab F4ac F4ad

A ● ● ●

B ● ●

C ● ●

D ●

E

F ●

G ●

H ● ●

1.5.2 ETEC F4ab and ETEC F4ac receptor

Early studies postulated the existence of three different receptors: bcd, which binds to all ETEC

F4 variants; bc, which binds only to ETEC F4ab and ETEC F4ac; and d, which binds only to ETEC F4ad

(Billey et al., 1998).

Page 20: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 19 -

A recent hypothesis suggests a one-locus model―a common receptor for ETEC F4ab and ETEC

F4ac (F4bcR) that is inherited as an autosomal monogenetic trait (Python et al., 2002; Jørgensen et al.,

2003)—or a two-loci model―two different receptors, but with closely linked loci for F4ab and F4ac

(Guérin et al., 1993; Peng et al., 2007).

1.5.3 ETEC F4ad receptors

The inheritance of receptors for fimbriae F4ad (F4adR) is not well understood. Bijlsma & Bouw

(1987) suggested a dominant receptor locus for F4adR that is inherited independently, but is closely

linked to the receptors for ETEC F4ab/F4ac. Enterocytes adhesive for ETEC F4ad have been found in

offspring of resistant parents, indicating a non-persistent inheritance of F4adR. The study postulated the

existence of an “intermediate” phenotype, caused by epistatic inhibitor genes, to explain the discrepancies

observed in F4adR inheritance. Hu et al. (1993) postulated, for ETEC F4ad, the existence of a high-

affinity receptor co-segregating with F4bcR and a low-affinity receptor expressed only in pigs under 16

weeks of age. In the high-affinity receptor, bacterial adhesion to the enterocytes is close to 100%, while in

the low-affinity receptor, the enterocytes can be either susceptible or resistant to ETEC F4ad in variable

percentages.

1.5.4 ETEC F4ac susceptibility among breeds

Among the three F4 fimbriae variants isolated in pigs with diarrhoea, F4ac is the most

predominant (Westerman et al., 1988). Susceptibility to ETEC F4ac varies largely among pig breeds.

European breeds, such as Large White and Landrace are highly susceptible with a rate of 40─88%

(Snodgrass et al., 1981; Edfors-Lilja et al., 1986). American breeds, such as Hampshire and Duroc, have

similar high susceptibility, with a rate of 46─62% (Baker et al., 1997; Yan et al., 2009)

Chinese breeds, such as Meishan and Fengjing, have been reported to be resistant to ETEC F4ac.

Minzhu and Songliao Black breeds have a low susceptibility, with a rate of 8─28% to F4ac (Chappuis et

al., 1984; Duchet-Suchaux et al., 1991; Michaels et al., 1994; Li et al., 2007). It is possible that, like wild

boars that are naturally resistant to ETEC F4ac, Chinese breeds have been isolated from the rest of the

world and have been subjected to disease pressure without strict selection by swine producers.

Page 21: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 20 -

1.6 Methods for preventing F4 diarrhoea

Several strategies are used worldwide to prevent diarrhoea caused by ETEC F4. One method is

to select resistant pigs for breeding by using genetic markers, such as SNPs or microsatellites, that exhibit

LD with F4bcR. The simplest methods of preventing E. coli infection are improving the condition of the

pigs, decreasing the density on the farms, having better hygiene and rooms with controlled temperature

and ventilation, and using prebiotics and probiotics in the diet. In addition, Lactobacillus seems to reduce

ETEC F4 adhesion to the intestinal cells (Blomberg et al., 1993; Roselli et al., 2007), and chelated zinc

and mannanoligosaccharides not only prevent diarrhoea in pigs, but are also growth promoters (Castillo et

al., 2008).

1.6.1 Antibiotics

Antibiotics used as growth promoters have been administered at low, sub-therapeutic doses for

long periods of time to control the microbial population of the intestine (NOAH, 2001). In the intestine,

almost 6% of the energy in the diet is lost due to microbial fermentation (Jensen, 1998).

However, pathogenic bacteria are becoming resistant against antibiotics or tolerant to high doses

due to the abuse as a growth promoter, thereby compromising their therapeutic use (Delsol et al., 2005;

Holt et al., 2011). The appearance of multi-drug resistant (MDR) phenotypes has led to the use of newer

antibiotics that are chemically similar to those used in human disease treatments (Rosengren et al., 2008;

Smith et al., 2010). Swiss legislation on animal nutrition has banned the use of antibiotics for growth

promotion in animal feed since 1999 in order to reduce antimicrobial resistance; the EU passed a similar

ban in 2006 (EFSA, 2007). Antibiotics used for disease prevention are administered at high doses for a

short period of time and are still used routinely on many farms.

1.6.2 Homeopathy

Homeopathic treatments have been studied as a replacement for the use of antibiotics in

livestock (Camerlink et al., 2010). Homeopathic treatments are highly-diluted preparations made with

Page 22: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 21 -

substances that produce symptoms similar to those of the disease being treated. The use of homeopathic

treatments on humans and animals has no scientific evidence of efficacy (ECH, 2005; Mathie, 2003).

In the Camerlink et al. (2010) study, pregnant sows were treated twice a week with a nosode, a

diluted preparation made from various strains of E. coli, during the last month of their pregnancies. The

homeopathic litters had slightly fewer cases of E. coli diarrhoea than the placebo groups.

1.6.3 Immune protection by colostrum and milk

Natural protection against ETEC F4 is provided by immunoglobulin type A (IgA), which inhibits

adhesion and colonisation of the gut (De Geus et al., 1998). During pregnancy in humans, primates, and

rodents, immunoglobulin type G (IgG) produced by the mother crosses the placenta (Pentsuk & Van Der

Laan, 2009). However, in cattle, sheep, horses, and pigs, there is no transplacental transfer of

immunoglobulins (Sterzl et al., 1966). The foetus is devoid of circulating antibodies until birth, and the

newborn is unable to produce its own immunoglobulins for several weeks and is vulnerable to bacterial

infections.

Piglets must absorb the immunoglobulins from the mother via colostrum, which contains high

levels of IgG, creating the first systemic immunoprotection against bacterial infection in the newborn

(Frenyó et al., 1981). Milk contains IgA, which is barely absorbed by the suckling pig, maintaining local

immunoprotection of the intestinal mucosa for the entire lactating period (Curtis & Bourne, 1971).

Effective immunisation against neonatal diarrhoea from F4 E. coli is achieved if piglets can

drink colostrum and milk from an ETEC F4 susceptible sow that has developed IgG and IgA antibodies

against the bacteria from previous infections. Resistant sows cannot be infected by ETEC F4; therefore,

they cannot produce antibodies to be secreted in the colostrum (Sellwood, 1979; Sellwood, 1982). As a

result, if newborns are susceptible to ETEC F4 they can develop neonatal diarrhoea.

In an experimental study, capsules containing anti F4 egg yolk immunoglobulins (IgY) were able

to reduce cases of diarrhoea in piglets (Li et al., 2009). IgY can be an alternative method of achieving

effective immunisation against E. coli in piglets.

Page 23: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 22 -

1.6.4 Vaccine

To protect newborn piglets, pregnant sows are vaccinated intramuscularly with an injection

containing F4 fimbriae or LT enterotoxins, so that the antibodies will be secreted in the colostrum and

milk (Jones & Rutter, 1972; Fürer et al., 1982). The maternal immunity helps to prevent diarrhoea during

the neonatal period, but it is lost during the weaning period. The lack of immunoglobulins from the milk

and the stress induced by the sudden diet changes make the piglets vulnerable to E. coli infections. Piglets

can be vaccinated with F4 antigens only during the weaning period; some vaccines will not work if

maternally-derived antibodies from the sow are still present in the piglets.

Vaccination can be achieved with purified F4 fimbriae, formalin-inactivated ETEC, engineered

bacteria containing the gene cluster expressing F4 fimbriae, or transgenic plants expressing fimbrial

subunit FaeG (Liang et al., 2006; Floss et al., 2007). Intramuscularly administered vaccines stimulate only

systemic immunity in piglets, blocking disease development only when the bacteria have crossed the

mucosal barrier of the intestine (McCluskie & Davis, 2000). Oral administration of antigens better

stimulates the production of IgA in the intestinal mucosa; however, the vaccine must resist the acid

environment and the digestive enzymes of the gastro-intestinal tract. The vaccine needs to cross the

intestinal barrier to stimulate the intestinal mucosal system and induce a protective immune response

instead of oral tolerance in the piglets (Van Den Broeck et al., 1999).

Antigens can be encapsulated to protect them from degradation (Galindo-Rodriguez et al., 2005).

In an experimental study, capsules made of methyl vinyl ether and maleic anhydride copolymer were able

to reduce cases of diarrhoea in piglets compared to non-capsulated antigens (Vandamme et al., 2011).

These vaccine treatments are still experimental and not yet commercially used on farms.

The vaccination procedures require the herding and restraint of the pigs and are quite stressful

for the pigs and time-consuming for the veterinarian or farmer. Furthermore, vaccines are expensive, not

optimal for stimulating intestinal immunity, and must be repeated for each new litter (Lee et al., 2001).

Breeding pigs for resistance to ETEC F4ab/F4ac adhesion is more economical, and the effects are lasting.

Page 24: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 23 -

1.7 Genetic mapping of F4bcR

An LD was observed between F4bcR and the transferrin gene (TF) by linkage analyses on three-

generation Wild Boar/Swedish Large White crossbreeds (Guérin et al., 1993; Edfors-Lilja et al., 1995). In

these studies, F4bcR was mapped to a 69 cM region on porcine chromosome 13 (SSC13) (Figure 1.4).

The F4bcR was refined by fine mapping in the interval between microsatellites S0068 and SW1030

(Python et al., 2002), and then to a 6 cM interval between microsatellites SW207 and SW225 (Python,

2003; Jørgensen et al., 2003).

Recently, a comparison of pedigree data from Swiss Large White and Landrace purebred pigs

and Large White/Landrace crossbreds have additionally refined the F4bcR locus in a 10 Mb interval (5.7

cM) between microsatellites SW207 and S0075 (Joller et al., 2009). Jacobsen et al. (2010), through

haplotype mapping the Swedish three-generation material from Edfors-Lilja, further refined the F4bcR

locus to a 3.1 Mb interval, between gene zinc finger DHHC type containing 19 protein (ZDHHC19) and

microsatellite S0075.

A transversion in intron 7 of the mucin 4 gene, MUC4_g.8227 G>C, co-segregates with the

F4bcR alleles (Jørgensen et al., 2004). This SNP is currently used by Danish breeding programs;

however, subsequent observations in Switzerland (Joller, 2009; Rampoldi et al., 2011) have raised doubts

as to whether this mutation in the MUC4 gene is useful for eliminating ETEC F4ab/F4c susceptible pigs

from herds, as this mutation is not in full LD with F4bcR. Recently, a patent application was filled for a

SNP in the mucin 13 gene (MUC13), transition A157G (named MUC13-813 in this study) (Zhang et al.

2008) that co-segregates with the F4bcR alleles (Huang et al., 2006). However, a subsequent study by

Ren et al. (2012) showed that the SNP, in some rare cases, is not in full LD with the F4bcR locus.

Other studies have investigated polymorphisms in candidate gene transferrin receptor (TFRC)

(Wang et al., 2007; Jacobsen et al., 2011), lactosylceramide 1,3-N-acetyl-beta-D-glucosaminyl transferase

(B3GNT5) (Ouyang et al., 2012), mucin 20 (MUC20) (Jacobsen et al., 2011; Ji et al., 2011), solute carrier

family 12 member 8 (SLC12A8), myosin light chain kinase (MYLK), karyopherin alpha 1 (KPAN1)

(Huang et al., 2008), non-receptor tyrosine kinase 2 (ACK1; TNK2 in this study), beclin-1 associated

RUN domain containing protein (KIAA0226), and MUC4 (Jacobsen et al., 2011). In all these studies,

haplotypes were associated with ETEC F4ab/F4ac susceptibility, but none of the polymorphisms were

causative.

Page 25: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 24 -

Figure 1.4: Location of candidate genes for F4bcR on SSC13. The gene order and the scale are deduced

from Sscrofa assembly 9, the positions of the microsatellites are shown to the right of the SSC13

ideogram, and the approximate positions of candidate genes are indicated on the right side (Rampoldi et

al., 2011 [modified version]).

1.8 Candidate genes for F4bcR

A candidate gene is a gene suspected of being involved with a particular disease or condition,

and it can be positional or functional. A functional candidate gene is involved in a specific function that

could be related to the condition investigated. In this study, functional candidate genes for F4bcR are

involved in the expression of receptors on the epithelial cell membrane. A positional candidate gene is

located in the chromosome area believed to be involved in the condition investigated. In this study,

positional candidate genes for F4bcR are located in the interval ZDHHC19-S0075 (Section 1.7) in the

reference sequence Sscrofa assembly 10.2.

Page 26: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 25 -

1.8.1 Positional candidate gene

1.8.1.1 SLC12A8

The solute carriers (SLC) are a group of membrane transport proteins that include over 300

members, organized into 51 families (Hediger et al., 2004). Solute carrier family 12 member 8

(SLC12A8) is a cation/chloride co-transporter. The human SLC12A8 gene (ENSG00000221955) contains

13 exons, possesses five isoforms, and encodes for a 714 amino-acid membrane protein of 78 kDa. The

SLC12A8 gene is conserved in chimpanzee, pig, dog, cow, mouse, rat, zebrafish, fruit fly, mosquito, and

C. elegans. It maps between 144625000 and 144760000 bp in SSC13 on the reference sequence 10.2. The

SLC12A8 protein exhibits low expression in normal skin, small intestine, stomach, testis, thyroid, and

colon. SLC12A8 protein may play a role in the control of keratinocyte proliferation, and it is a candidate

gene for psoriasis susceptibility in humans (Hewett et al., 2002; Hüffmeier et al., 2005). In pigs, SNP

SLC12A8_159 A>G has been shown to be in LD with F4bcR (Huang et al., 2008).

1.8.2 Positional and functional candidate genes

1.8.2.1 HEG1

Heart of glass (HEG) is a regulating protein for heart growth in zebrafish (Mably et al., 2003).

HEG homolog 1 (HEG1) protein is an orphan receptor in mammals, related to the mucin family (Lang et

al., 2006). The human HEG1 gene (ENSG00000173706) possesses two isoforms and encodes for a 1381

amino-acid protein of 147.5 kDa with calcium-binding EGF-like domains. These domains are composed

of 30─40 amino-acids with six cysteine residues involved in three disulphide bonds (Downing et al.,

1996; Bork et al., 1996) and are normally found in extracellular proteins. The HEG1 gene has been

reported to be expressed in human, chimpanzee, cattle, mouse, and pig. It maps between 144760000 and

144830000 bp in SSC13 on the reference sequence 10.2. A recent genome-wide association study on 301

Landrace, Yorkshire, and Songliao Black piglets showed a possible LD with F4bcR (Fu et al., 2012).

Page 27: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 26 -

1.8.2.2 MUC13

Mucins are glycoproteins on the membranes of mucosal epithelial cells (Williams et al., 2001).

They form gels and are used for lubrication, cell signalling, and forming chemical barriers (Marin et al.,

2008). Mucins contain EGF domains and oligosaccharide structures used as binding sites for bacteria.

Mucin 13 (MUC13) is a transmembrane glycoprotein involved in cell cycle regulation. The human

MUC13 gene (ENSG00000173702) encodes for a 511 amino-acid protein of 54 kDa. MUC13 protein is

highly expressed in a variety of epithelial carcinomas (Maher et al., 2011). The MUC13 gene maps

between 144992000 and 145021000 bp in SSC13 on the reference sequence 10.2. Quantitative PCRs

have shown no difference in gene expression between pigs resistant and susceptible to ETEC F4ac

adhesion (Schroyen et al., 2012). A recent study by Ren et al. (2012) showed two isoforms of MUC13 in

pigs, MUC13A and MUC13B, each with distinct tandem repeats in exon 2. The repeats in MUC13B

encode for a heavily O-glycosylated region in the protein, a possible binding site for ETEC F4ab/F4ac.

The O-glycosylation binding site is not present in MUC13A. Pig SNP A157G (SNP MUC13-813 in this

study) has been shown to be in LD with F4bcR and was recently patented as a marker for selecting pigs

resistant to ETEC F4ac (Huang et al., 2006).

1.8.2.3 ITGB5

Integrins are protein receptors that mediate attachment between cells or between a cell and the

extracellular matrix (ECM). Integrins give information from the ECM to the cells and reveal the status of

the cells to the outside, enabling rapid responses to sudden changes in the environment. Integrins are

involved in several biological activities, such as immune patrolling and cell migration, and they can be

used by viruses for binding to cells. The integrin β-5 (ITGB5) protein is a receptor for fibronectin. The

human ITGB5 gene (ENSG00000082781) encodes for a 799 amino-acid protein of 88 kDa. The ITGB5

gene maps between 145042000 and 145114000 bp in SSC13 on the reference sequence 10.2. Recent

studies have shown a possible LD with F4bcR (De Greve et al., 2007; Shahriar et al., 2006; Fu et al.,

2012). In pigs, SNPs ITGB5_c.920 C>T, ITGB5_c.1580 G>A, ITGB5_c.1715 C>T, ITGB5_c.2744 G>A,

ITGB5_g.31420 C>A, and ITGB5_g.31487 G>A were shown to be in LD with F4bcR (Huang et al.,

2011).

Page 28: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 27 -

1.9 Objectives

The aims of this study were threefold:

1) Analyse the porcine sequence in candidate genes for differences that are associated with ETEC

F4ab/F4ac adhesion phenotypes in informative families.

2) Develop a reliable genetic test that discriminates between pigs susceptible and resistant to ETEC

F4ab/F4ac.

3) Produce informative matings to elucidate the inheritance of the F4ad receptor (F4adR) allele(s).

Page 29: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 28 -

2. MATERIALS AND METHODS

2.1 Pigs

The pigs used in this study originated from a university experimental herd (UEH) at the

Department of Farm Animals, Faculty of Veterinary Medicine, University of Zurich. The UEH was

studied in several papers in collaboration with Jørgensen’s group from the Department of Basic Animal

and Veterinary Sciences, University of Copenhagen, Denmark, to identify the receptors for E. coli

F4ab/F4ac adhesion (F4bcR) and the F4ad fimbrial receptor (F4adR) inheritance mechanisms (Joller et

al., 2009; Jacobsen et al., 2010; Rampoldi et al., 2011).

In 1998, a Large White purebred family and a Large White/Landrace crossbred family were

originally bred for eight generations. In the UEH 249 matings were performed, generating 2565 offspring.

Since the year 2000, the E. coli F4 microscopic adhesion test (standard MAT), performed on one

intestinal site, was used routinely to determine the inheritance of the resistant and susceptible alleles to

the three different variants of E. coli F4 fimbriae; 2372 pigs were phenotyped with the standard MAT.

Since the year 2009, together with the standard MAT, 537 pigs were tested in four intestinal sites to

determine the F4ad fimbrial receptor phenotype (F4ad MAT).

Jørgensen’s group provided haplotype information based on 10 Nordic Experimental Herd

(NEH) pigs and 10 Swiss pigs used in this study for selecting SNPs and candidate genes to investigate.

This study also performed analyses on a representative sample of the Swiss porcine population,

initially with 78 pigs from 38 litters from the Landrace and Large White breeds, randomly selected at the

Swiss Performing Station of Sempach (SPS) (Joller, 2009), and later with 40 other pigs that were

randomly selected to test newly discovered markers.

Page 30: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 29 -

2.2 Determination of the ETEC F4 receptor phenotype

2.2.1 Preparation of ETEC F4 strains

The Escherichia coli F4 strains E68I (O141:K85ab:F4ab), G4 (O45:K(E65):F4ac), and Guinée

(O8:K87:F4ad) (Thorns et al., 1987) were obtained from the Veterinary Laboratories Agency Weybridge,

Surrey, GB. Stock bacteria from confluent growth on agar plates were harvested and frozen at -70°C in

0.5 ml trypticase soy broth (TSB) containing 10% glycerine. Each batch of frozen stock suspension was

confirmed by slide agglutination with OK antisera. The antisera were produced at the Institute of

Veterinary Bacteriology, University of Zurich.

Bacteria were grown on Columbia Sheep blood agar plates (Appendix) and stored at 4°C. Every

second month, material was taken from five colonies and plated on fresh agar plates. After three transfers

to blood agar plates, the cultures were renewed from frozen stock. After each transfer to new blood agar,

the expression of F4 fimbriae bacterial subculture was confirmed by slide agglutination of confluent

growth with polyvalent F4 antiserum or by F4 strain-specific PCR (Table 2.1) (Alexa et al., 2001). The

antisera were provided by the Bundesinstitut für Risikobewertung (BfR), Berlin, Germany.

Colonies from confluent growth from the blood agar plates were picked and grown at 37°C for

24 h in TSB (Appendix) test tubes one day before use. Shortly before use, 1 ml of bacterial culture was

diluted 1:10 in pre-warmed TSB and incubated at 37°C for 90 min to achieve maximal density of the

culture and maximal fimbriation of the bacteria.

2.2.2 Sampling of intestinal tissues

The pigs were slaughtered routinely, at the age of two months, at the age of seven months (~ 100

Kg), or when they were eliminated from breeding. Usually, feed was withheld for 16 hours before the

pigs were slaughtered; water and straw were always offered. Blood samples and tissue samples of the

intestine were taken at the time the pigs were slaughtered. The interval between exsanguination and

sampling of intestine was between 20 and 40 min.

For the standard MAT, starting at the cranial mesenteric artery, the intestine was separated from

the mesentery, and a 10─20 cm empty segment of jejunal intestine was taken anywhere between 3.5 and

Page 31: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 30 -

7.5 m distal from the cranial mesenteric artery. For the F4ad MAT, four segments were usually taken: one

was taken 2 m distal from the cranial mesenteric artery (A); another one 2 m proximal to the ileocaecal

valve (D); and the other two segments at 1/3 (B) and 2/3 (C) of the distance between A and D. All

segments were opened longitudinally, placed in wide-necked bottles containing 80 ml of 4°C PBS-

EDTA, and stored at 4°C until further processing.

Intestinal samples for RNA extraction were taken after the pigs were slaughtered. Intestinal

scrapings were removed with glass slides from an intestinal segment free of contents. The scrapings were

wrapped in aluminium foil or put in 1.5 ml tubes and frozen in liquid nitrogen. Samples were stored at

-70°C until RNA extraction.

2.2.3 Preparation of enterocytes

The enterocytes were prepared for the MAT according to Sellwood et al. (1975), with

modifications of Vögeli et al. (1996) and Python et al. (2002). All four segments were tested for F4ad,

while segment B or C usually was tested for F4ab and F4ac.

The superficial layer of the intestinal segment was scraped off the surface with a microscope

slide and collected in 50 ml centrifuge tubes containing 30 ml of PBS-formaldehyde. The suspension was

stirred vigorously with forceps for 1 min and stored at 4°C for 15 min, letting large tissue fragments

sediment. The supernatant was decanted and stored at 4°C again, for 20 min, to sediment the remaining

large tissue fragments. Subsequently, the supernatant was centrifuged at 200 g for 10 min, and the pellet

was carefully resuspended in 10 ml of PBS and centrifuged again. The enterocytes were resuspended in 5

ml mannose buffer and diluted to a concentration, judged by eye, of 105─106 cells/ml.

2.2.4 Microscopic adhesion test

One millilitre of resuspended enterocytes was incubated in a 6-well macroplate at 37°C for 30

min with 1 ml freshly grown culture from each of the three ETEC F4 strains. Subsequently, 20 well-

separated and intact enterocytes were scored for each sample under a light microscope with 400x

magnification.

Page 32: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 31 -

Adhesion strength was expressed as the percentage of adhesive enterocytes. An enterocyte was

classified as adhesive if more than five bacteria adhered to the brush border (Figure 2.1). Twenty

additional enterocytes were scored if adhesion of more than five bacteria were observed in >0%─30% of

the scored enterocytes. Initially, pigs with more than 15% E. coli F4ac adhesive enterocytes and pigs with

more than 2.5% F4ab and F4ad adhesive enterocytes were considered to be susceptible. Changes in

classification were made for F4ad adhesive enterocytes, as described in Section 3.8.1. The same person

performed the jejunum sampling and purification and classification of the enterocytes.

Figure 2.1: Determination of the ETEC F4 receptor phenotype in the MAT after preparation of

enterocytes. Cell without adhesion (top left) and cell with multiple ETEC F4 adhering to the brush border

(bottom right) (Python, 2003).

2.2.5 Positive and negative control of bacterial adhesion

Beginning in July 2006, adhesive and non-adhesive enterocytes were kept for further use as

positive and negative controls for F4 adhesion. After scoring, the remaining enterocytes in mannose

buffer and the cells of the second decantation were pooled, supplemented with 10 ml of PBS, and stored

at 4°C for 5 min for sedimentation of large tissue fragments. After a centrifugation step at 200 g for 10

min, the pellet was resuspended in 5 ml of DMSO-Hanks medium (Bosi et al., 2004). Aliquots of the

suspension were frozen in cryotubes at -70°C.

Page 33: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 32 -

Control cells were included in each test series. Before use, the cells were thawed at room

temperature, diluted in 10 ml of PBS-formaldehyde, and centrifuged. The pellet was washed in PBS,

centrifuged, resuspended in 1 ml of mannose buffer, and was ready for incubation with bacterial strains.

2.3 DNA methods

2.3.1 DNA extraction from blood

Blood was collected in Vacuette or Venosafe tubes containing EDTA and stored at 4°C or 20°C

until processing. DNA extraction from blood samples was performed using a lysis method, as described

by Vögeli et al. (1994). In brief, 600 µl of blood were mixed with 500 µl of lysis buffer (Appendix), left

at room temperature for 15 min, and then centrifuged at 13,000 g for 30 s. The pellet was resuspended in

1 ml of lysis buffer, vortexed, and left at room temperature for another 15 min. The mixture was then

centrifuged at 13,000 g for 30 s. The resuspension and centrifugation were repeated two more times. The

pellet was resuspended in 200─400 µl of PCR turbo buffer, and 20─40 µl of Proteinase K (20 mg/ml)

were added to the suspension. After incubation at 55°C for 2 h and deactivation of the Proteinase K at

95°C for 10 min, the samples were stored at -20°C until further use.

As an alternative, a column-based purification for the DNA was made with the GenElute

Mammalian Genomic DNA Miniprep Kit (Sigma-Aldrich); 500 µl of blood were mixed with 1400 µl of

cold ECL buffer, then centrifuged for 10 min at 13,000 g. The washing step was repeated one more time,

and the pellet was resuspended in 200 µl of resuspension solution from the kit. The next steps followed

the white blood cell (WBC) preparation protocol of the kit. After the elution of the DNA from the

column, the samples were stored at -20°C until further use.

The DNA concentration was measured using a Qubit® (1.0) fluorometer with a Qubit® dsDNA

HS (high sensitivity) assay kit (Invitrogen). Alternatively, a NanoDrop ND-1000 spectrophotometer

(Thermo Scientific) was used.

Page 34: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 33 -

2.3.2 Polymerase chain reaction

2.3.2.1 Primer design

Primers for PCRs and sequencing were based on porcine DNA or RNA sequences or on

homologous and conserved DNA sequences of Homo sapiens published on the EMBL/GenBank

databases. Primer sequences were usually 20─24 bp in length, with a GC content of 40─60% and one or

two G/C clamps at the 3’ end. The primers were designed with the web-based software Primer3 v. 0.4

(Rozen & Skaletsky, 2000; http://frodo.wi.mit.edu). All the primers were tested for hairpins and self- and

hetero-dimers with the web-based software Oligo Analyzer v. 3.1 (Owczarzy et al., 2008;

http://eu.idtdna.com/analyzer/Applications/OligoAnalyzer/default.aspx) (Tables 2.1, 2.2.1, 2.2.2, and

2.5). Primers for pyrosequencing were designed with Pyrosequencing Assay Design v. 1.0 software by

Biotage (Table 2.3).

2.3.2.2 Standard PCR

PCRs were generally performed in a 25 µl reaction volume containing 50─250 ng of DNA, 200

µM of each dNTP, standard PCR buffer (10 mM Tris-HCl pH 8.3, 50 mM KCl, 1.5 mM MgCl2, 0.001%

gelatine), 10 pmol of each forward and reverse primer, up to 0.5 mM additional MgCl2, and 1.5 U Taq

DNA polymerase or 0.75 U Taq DNA JumpStart polymerase. Amplification was carried out in 200 µl

single tubes, 8-tube strips, or 96-well plates on a PTC100 (MJ Research, Bioconcept, Allschwil) or a

TPersonal thermocycler (Biometra, Biolabo, Châtel-St-Denis). After an initial denaturation at 94°C for 2

min, the samples were cycled 30─38 times, as follows: denaturation at 94°C for 30 s, annealing at

50─65°C for 30 s, and extension at 72°C for 30─45 s. At the end, the samples were extended at 72°C for

5 min (Tables 2.1 and 2.5).

Page 35: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 34 -

Table 2.1: Primers for standard PCR are given with the SNPs, DNA fragment size, annealing temperature,

and position of SNPs or amplicon according to the reference sequence Sscrofa 10.2.

Gene

Primers Forward and Reverse

Annealing

temperature

(°C)

Amplicon

length

Position on

Sscrofa

10.2 Position of SNPs

F4ab1 K88ab-F: TTGCTACGCCAGTAAGTGGT

K88ab-R: CGAAACAGTCGTCGTCAAA 65 296 ---

F4ac1 K88ac-F: TTTGCTACGCCAGTAACTG

K88ac-R: TTTCCCTGTAAGAACCTGC 54 436 ---

F4ad1 K88ad-F: GGCACTAAAGTTGGTTCA

K88ad-R: CACCCTTGAGTTCAGAATT 50 169 ---

ACTB2 BACT-F: TCCCTGGAGAAGAGCTACGA

BACT-R: CGCACTTCATGATCGAGTTG 54

250

cDNA

150

---

TNK2

g.7075 TNK2e6-7-Fc2: GGAACCACTTTGATTGTTCTC

TNK2e6-7-R: GACAGGGACACCAACAGCTAA 61 625 143654201

g.7717 TNK2e9j-F: GAAGAGCTGGGTGCTCTGCTT

TNK2e9j-R: AGCGGCAGCTGCATACTTGA 61 551 143654843

g.11142 TNK2e12b-Fv2: CCTGTGAGTGAAGACCAAGACC

TNK2e12b-Rv2: CCCCATCTCCTCCTCACTTGA 61 751 143663677

MUC4

g.6242; g.6308;

g.6317; g.6321;

g.6609; g.6616;

g.6634; g.6675;

g.6690; g.6745;

g.6770; g.6862

MUC4-6136-F: GTTACTGGCCTCGACTCTCC

MUC4-6887-R: AGGTTGTACCCTTGGCATTC 61 749

143818524-

143819273

g.7947 MUC4-7741-F: GGTCCTACGCCTTGTTTCTC

MUC4-8202-R: CCTTCATGGGGTTGTTGTAATA 61 462 143820333

g.8227 MUC4-8012-F: CACTCTGCCGTTCTCTTTCC

MUC4-8378-R: GTGCCTTGGGTGAGAGGTTA 56 367 143820613

LRCH3

212; 213; 214;

215; 2163

LRCH3-F1: TGCCTGACATTTTGCTAACG

LRCH3-R1: CTGCACTTGTGGTGGAGAAC 52 643

144104687-

144105329

217; 218; 2193 LRCH3-F2: TTGAGGAGAGTTGCATGTTGTT

LRCH3-R2: TCCTGCTCAGTGGATTAAAGG 52 405

144109773-

144110177

LMLN

g.15920 LMLN-I4F: GGCACTATCTTACTTAGCAG

LMLN-I4R: TGGTTTGTTGCACATTGT 50 528

144201963/

144255122

ZNF148

g.96828 ZNF-INF1: TGTGCTTTAACCCTTATACTCTGC

ZNF-INR1: TCTTCATTCATACAGGTATTTCTTGA 53 668 144544700

SLC12A8

SLC-C1F: GCCCAGATGTCTCAAGTGCAGG

SLC-C1R: GACTCTCCGGGGCTGTAATCTG 60

cDNA

690

144499474-

144717264

Page 36: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 35 -

Continued from previous page

Gene

Primers Forward and Reverse

Annealing

temperature

(°C)

Amplicon

length

Position on

Sscrofa

10.2 Position of SNPs

SLC12A8

SLC-C2F: GCCATGTACATCACCGGCTTTGC

SLC-C2R: TGGCCTTCTTGGTCTTCCCCTTC 61

cDNA

1087

144644897-

144751894

SLC-C3F: CATCGCGGTGGATTACTCTT

SLC-C3R: TGCATATGCGGGTGTAGAAA 53

cDNA

742

144751590-

144861506

g.1365 SLC-D4F: CCTAACCTGGGAACCTCCAT

SLC-D4R: GCCTTGTCTGGTTGAGAGGA 57 335 144499531

g.22157 SLC-D3F: CTGTGAAGAGGCTGGGAAAG

SLC-D3R: TGTGGGATACACAAGGGTGA 57 457 144631142

1594 SLC-E10-11-F: AGAAATACACAATGGCAGCAA

SLCE10-11-R: TTCCTAAAGTCCAGGCTCCA 53

728

cDNA

177

144862026

g.129502; c.1990 SLC12A8-13F: AAAACCTCTGAGGGCCAGAT

SLC12A8-13R: CACACCACAATTCACCCAAG 57 677

144853837-

144853159

SLC-C4F: CTCCCAAGAGCAGATCATTTTG

SLC-C4R: CATAACAAAATGCGGGGAGTAT 54 641

144849975-

144849335

c.2947 SLC-C5F: TTTGTTATTGTCATGGTTCCAAAC

SLC-C5R: ATTAAAAAGGCCTTCACTAAACAA 51 521 144849048

HEG1

g.5244 HEG1-F: CCCATCGTAGCTCACAGG

HEG1-R: CCTCCACGTCTGCATGTG 57 353 144823489

c.321 HEG-E2F: GCATCTCCTTCTCCACTGGTAT

HEG-E2R: AGCTATAGCAAGCCACAGGTGA 57 569 144800514

HEG-E5F: TGGCCTTACATCTTCCAAGG

HEG-E5R: GGCTGAAACGTAGGTGTGGT 53

cDNA

1131

144791443-

144783842

HEG-E5F1: TGTACCCCCTGTAGTGGATTTT

HEG-E5R1: CCTATGTGGAGACTCACTCAAGT 55

799

144790922-

144790128

HEG-E5F2: ATTGTGGCTCAGTGGGTTTAAG

HEG-E5R1: CCTATGTGGAGACTCACTCAAGT 55 895

144789427-

144788577

HEG-NEF: GTTCTGAATCGGAGGAAGGAG

HEG-NER: GAGAATGTGCAAAAAGATGCTG 54 287

144784843-

144784557

c.1905; c.1917 HEG-E6F: ACCTACGTTTCAGCCCCTTT

HEG-E6R: GCTTGGGAGCTTTGATTCTG 55 696

144783856-

144783161

g.49379 HEG-E7F: TTTCCTGGGTGATTAACTGCTT

HEG-E8R: TGCAAATAAAGTGGAGGAAGAA 52 735 144779899

HEG-C1F: TTCCACAGCTGAAGTGGTGA

HEG-C1R: CTGCAGGCATTGACACATTT 53

cDNA

603

144782675-

144771912

HEG-C2F: ATGCTTCTAGGGAGCCCAGT

HEG-C2R: TTCGTTCAAGTTCGGGATTC 53

cDNA

684

144772271-

144704755/

144928236

c.6955 HEG-INF: TGTTAAGCTGACTCTCTCATGCTC

HEG-INR: CACTCCTGCCTGAGGTAAAAATAC 57 408

144707761/

144931242

Page 37: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 36 -

Continued from previous page

Gene

Primers Forward and Reverse

Annealing

temperature

(°C)

Amplicon

length

Position on

Sscrofa

10.2 Position of SNPs

HEG1-MUC13

(JN613413)

23529 MCL23-F: CCACGAAATACCATTCAGCA

MCL23-R: GTTGAACTGGGAATCGAACC 53 547 144935458

34623; 34996;

35253; 35314

HEG1-24F: CCCTGGACCTTTATCCCAAC

HEG1-24R: ATGGAGCTTTGAGGCCATTA 53 781

144946486-

144947266

53252; 53277;

53373; 53494;

53612

HEG1-22F: CTGGGGAGACAACCTACCAA

HEG1-22R: CCATTGGTGGATGGGTAAAC 55 472

144965707-

144965236/

160909987-

160910454

61641; 61768;

62196

HEG1-25F: CAGCAAGCATCGTCATTTTC

HEG1-25R: TGGGAACGTCCTCCAATAAA 53 699

144973759-

144974381

MUC13

g.207 MUC13-MF: TTGGCGAGGAAATCTACAAAAT

MUC13-MR: GGGAAATCATTTTCTCTCTGGA 54 221 144986614

g.791; g.1248;

g.1345

MCL25-F1: GAGTAGGGATCTGGTTTGATGC

MCL25-R1: GCAGAATTTGGCTCTTCACC 55 675

144987144-

144987818

g.1412; g.1414;

g.1945; g.1974

MCL25-3F: AAGAAAACTGGCTTATGAATGAT

MCL25-3R: TGTCAATCTGAGTTTAATATTCCTC 51 815

144987753-

144988561

g.8951; g.8981 MUC13-E2F: AAAGCATGTTTCTGGGTGCT

MUC13-E2R: AGTGTGGATCCCCAAAATGA 53 745

144996366-

144997115

c.232 MUC13-E3F: GCGTGTATGTGTAACTGGGTCA

MUC13-E3R: TGGGGTCATGTTTTTCCTATTC 54 394 144997797

g.15150 MUC13-B2F: GGGGAGCTCTGCATTGTATC

MUC13-B2R: TACAAAGAGGGGGAAACGTG 55 208/162 145003209

g.15376;

g.15379; g.15381

MUC13-E4F: ACCATGTGTGTAAGTCGCTGAG

MUC13-E4R: ACGTTTCCCCCTCTTTGTAGTT 55 361

145003308-

145003668

227; 226; 225;

224; 2233

MUC13-F: TGAGCAAGATGAGTGCCCCAGT

MUC13-R: TAGCCAGGCAGGCACAAGCA 58

536

cDNA

186

145010233-

145010768

813; 814; 829;

895; 905; 908;

920; 933; 9355

MUC13-7F: ATGTGGAAGAACAGAACTTGATTGAG

MUC13-7R: ATAGTCAGGGCGGGGTATACTACC 59 176

145016886-

145017061

c.1243; c.1289;

c.1290; c.1702

MUC13-C2F: AGCATGCTCTGATACCTGCAATGC

MUC13-C2R: TCCTTCCTGAAAGCTGGGAGACAT 63 1061

145010679-

145019845

c.1788; c.1842;

c.1930; c.1986;

c.2014; c.2068

MUC13-ED2F: CAGGGAAGGCTGAGACTTTG

MUC13-ED2R: ATGTCTCCCAGCTTTCAGGA 57 696

145019822-

145020517

MUC13-ITGB5

(JN613413)

107599; 107649 MUC13-3AF: ACACATTTTTGGCTGGTTCC

MUC13-3AR: GGATGCTCTCACCTGCAAA 53 717

145020893-

145021609

Page 38: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 37 -

Continued from previous page

Gene

Primers Forward and Reverse

Annealing

temperature

(°C)

Amplicon

length

Position on

Sscrofa

10.2 Position of SNPs

MUC13-ITGB5

(JN613413)

115242; 115364;

115460

M13IT-F3: TTTCAGGCTTGTTTTCCTCAA

M13IT-R3: TCAGCTGTGTTACCGAATGC 51 774

145028422-

145029195

ITGB5

c.246 ITGB5-E2F: GCTCAGCTTCCCTCATGAAC

ITGB5-E2R: GGGGTGGGGTTTATCCTCT 57 389 145064038

g.65464; c.917;

c.9206

ITGB5-E5F: GTCCCTAGCCCTCACCATCT

ITGB5-E5R: AGTCAGGCTGGGTCTCTCCT 59 363

145108252-

145107890/

160895287-

160894925

g.115393 ITGB5-F: CTTGGGGTAGAGGAAGTTGATG

ITGB5-R: CAGGTTGCTGAGACAGACTTTG 57 725 ---7

c.1580; c.17156 ITGB5-E9F: AGGTAACGGGAGTCCCAGAC

ITGB5-E9R: AGGACCATGGCAGTTGGTAG 57 588 ---7

c.27446 ITGB5-E14F: CCTCCAACCCTGTGTGAACT

ITGB5-E14R: AGGAACGGGCACGTCATAA 55 667 ---7

1 Alexa et al., 2001. 2 NCBI accession no. AJ312193. 3 Jacobsen et al., 2010. 4 Huang et al., 2008.

5 Zhang et al., 2008. 6 Huang et al., 2011. 7 SNPs were not found in reference sequence Sscrofa 10.2.

SNPs in the intergenic regions HEG1-MUC13 and MUC13-ITGB5 were named according to the position

in BAC clone CH242-101B19 and its accession no. JN613413 on NCBI. All the SNPs are enlisted

according to their physical positions in the genomic DNA, which sometimes do not correspond with their

supposed mapped positions in SSC13 due to errors in the reference sequence 10.2.

2.3.2.3 Long-range PCR

Standard PCR was used for DNA fragments up to 1000 bp. To amplify longer fragments, the

SequalPrep™ Long PCR with dNTPS kit (Invitrogen) was used. This kit was designed to amplify DNA

fragments of up to 15 kb in length, for use in next-generation sequencing applications. The kit included

all components for PCR except template, primers, and water. The long-range PCRs were carried out in a

20 µl reaction volume containing 2 µl of SequalPrep™ 10 X reaction buffer (which included MgCl2 and

dNTPs), 0.4 µl of DMSO, 1 µl of SequalPrep™ 10 X Enhancer (A or B), and 0.36 µl of SequalPrep™

Long Polymerase (5 U/µl). The DNA and primer amounts were the same as in the standard PCR. In GC-

Page 39: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 38 -

rich DNA samples, betaine was added to the master mix (1 M final concentration) in order to improve the

PCR (Chen et al., 2004b).

Amplification was carried out in 8-tube strips or 96-well plates. After an initial denaturation at

94°C for 2 min, the samples were cycled 30─40 times, as follows: denaturation at 94°C for 10 s,

annealing at 56─60°C for 30 s, and extension at 68°C. The extension time depends on the amplicon

length, generally 1 minute/Kb. After the first 10 cycles, the extension phase was prolonged for 20 s in

each cycle. At the end, the samples were extended at 72°C for 5 min (Tables 2.2.1 and 2.2.2).

Table 2.2.1: Primers for long-range PCR are given with the annealing temperature, expected DNA

fragment size, and position of the amplicon according to the reference sequence Sscrofa 10.2. These

primers were used to amplify the intergenic region HEG1-MUC13.

Primers Forward and Reverse Annealing

temperature (°C)

Amplicon

length Position on Sscrofa 10.2

HEG1-MUC13

L1F: GTAATGTAGCTTGGGACATGAAGGT

L1R: CTCTAGGAACCATCAGATCCATCA 58 7823 144933006-144940829

L2F: GCCACAGTTATCTTAATTCCCTGTC

L2R: GACAAGCAATGAGGTCCTTCTCTAT 60 8023 144940638-144948661

L3F: ACCCCACAGTGATACAGATATAACC

L3R: AGACCAGCCACTAGAAGTGAAGTAG 57 7914 144948335-144956354

L4F: GTATTTTGGGAGTGTTTCCTCCTAT

L4R: GACCACCAATCTACTTTCTGTCACT 57 7838 144956163-144964000

L5F: AGCATAGAATTACCATGAGATCCAG

L5R: AATACCTAACAACAGGAGAACAACG 56 8055 144963639-144971845

L6F: GAGAGTGTTTCAAAGTCAGGTCTTC

L6R: ATTCCCTTCTAGGACTTTACCCTAAG 58 7825 144971547-144979371

L7F: GATTTGTCCAGAGATAAGCCTCAAT

L7R: AAACAATGGTGTAGAGAGGAGAACC 57 8486 144979097-144987582

AF: TTTCTCCAGATATATGCCAAGAGTG

AR: CCATCTCCAAGATACACAATGCTT 56 5722 144978702-144984424

BF: ATAGGATTTGAGACTCTGCAGCTAA

BR: TTGAGGCTTATCTCTGGACAAATC 56 5015 144982886-144987900

L8F: CTGTGATGAAAGCAGGTAGAACATT

L8R: GATACTAGTTGGGTTCTTAACCTGTTG 60 7212 144986968-144994180

L9F: CCACCTCAGGTAAGAGAGATTCC

L9R: GTACCACCCGAATATCAGTCTAACA 57 7219 144992780-144998686

Page 40: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 39 -

Table 2.2.2: Primers for long-range PCR are given with the annealing temperature, expected DNA

fragment size, and position of the amplicon according to the reference sequence Sscrofa 10.2. These

primers were used to amplify the intergenic region MUC13-ITGB5.

Primers Forward and Reverse Annealing

temperature (°C)

Amplicon

length Position on Sscrofa 10.2

MUC13-ITGB5

L1F: CTTCATTCCATGTAACCCAAGTTC

L1R: AGCCAAGCCCTTTTAGTTAGTTGT 56 8283 145028579-145036861

L2F: ATTCTTTGGGTCTCTCTGAGTGTGAT

L2R: GCTGGAGTAAACACACAGATGAAAG 58 8039 145036439-145044477

L3F: ATTCAACAGTACCTAGAGACCCAAC

L3R: GAGTTTGTTTCCGTCTTGCTACTA 56 8285 145044740-145053024

L4F: CACAAAGGCTATTTTTGATGGACA

L4R: GTTGTGCAGAAAAAGTGTGCAGAT 56 8446 145052603-145061048

L5F: GGCAAAGGAGAAGACACAAAAGTAT

L5R: CAGGAGTAAAGAGCAATAAGGGAAA 56 7863 145060619-145068481

L6F: TCCTTAACTACGAAATGACAACAGG

L6R: AATGGTCTGTCACAGGTAGCAATAG 56 5629 145068329-145073958

L7F: AAGGAGAAGAAGGACTAAACATCCA

L7R: TGAACATTCCTCTGTTTGAGTCAG 56 8359 145073682-145082120

L8F: AAAAGTCTGTCTGAAGGATATGGAG

L8R: GTGTTGGAGCTTCTCATTCTAAACT 56 8393 145081837-145090229

L9F: CAGGAAATTACTGGGCAAGAGAATA

L9R: GACTCTCTAGAAGTTGCCTCTGAGTT 56 8681 145089847-145098527

L10F: AGGAAGAGAAAGGGTTTGAAGAGAA

L10R: TCTTTTAATGAGCCGTTCTGATCC 56 8420 145098283-145106702

L11F: AGGGTAGGAAAAGAAACACTGACTT

L11R: GTAGGCTTTTTAAGGTCTGGAACA 56 5439 145106518-145111956

2.3.2.4 PCR for pyrosequencing

The PCRs for pyrosequencing were performed in a 40 µl reaction volume. The amount of DNA,

dNTPs, PCR buffer, primers, MgCl2, and Taq polymerase were comparable to a standard PCR. One of the

primers was biotinylated at the 5’ end. Amplification was carried out in 96-well plates. After an initial

denaturation at 95°C for 5 min, the samples were cycled 45─50 times, as follows: denaturation at 95°C

for 15 s, annealing at 53─59°C for 30 s, and extension at 72°C for 15 s. At the end, samples were

extended at 72°C for 4 min. These PCRs were designed to amplify fragments no longer than 300 bp

(Table 2.3). The sequencing primers annealed to the opposite strands of the biotinylated primers.

Page 41: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 40 -

Table 2.3: Primers for pyrosequencing are given with the SNPs, DNA fragment size, and annealing

temperature. The biotinylated primers are written in bold.

Gene SNPs

F: forward-, R: reverse-,

S: sequencing primers

Annealing

temperature (°C)

Amplicon

length Position of SNPs

ZDHHC19

g.4043 T/C

F: TCCTACGAGGGCAAGGTATGTG

R: GAACTGAATCTGGTCTGGAATAGC

S: GCAAGGTATGTGGGGA

53 171

KIAA0226

g.62250 A/G

F: GCGGGAGCTGCTAGCCAA

R: GGGCTTCGGTGGGTTCCT

S: CCTCGTAGTCCGACA

59 70

MUC13

813 T/C F: AAAGAGGTTTCCTGCTCCTAGGT

R: GCCATTGTCAGATCCTAATTCC

S: TTGCAGGTTTTGAAAGT

55 148 814 G/T

ALGA0072075 C/T

F: TCGTTTTCATCTCTGGCAGATTG

R: CAGCCACTCAGGTCTCCATACTCT

S: CATGCGTTGGAGAAT

56 105

ALGA0106330 A/G

F: TTGGGCCCCATCTTTGGA

R: CCTCCCCTTTGACTGTCACATCT

S:AGGTAGCTGAGCCCC

55 184

2.3.3 Agarose gel electrophoresis

The PCR products were run on 0.8─1.5% agarose gels in 0.5 X TBE buffer containing 100 µg/l

EtBr at 75─100 mA for 0.5 to 1 h. A 6 X DNA loading dye was added to the PCR products prior to

loading. Depending on the size of the fragments, a 50 bp, 100 bp, or 1 Kb size standard was used. DNA

was visualized after the run under UV lights.

2.3.4 Genescan analysis

Microsatellite polymorphisms of labelled PCR products were visualized by genescan analysis

(Table 2.4). A mixture of 0.5 µl PCR product, 2.5 µl formamide, and 0.5 µl genescan 350 size standard

was denatured at 95°C for 5 min, and 1─2 µl were loaded on a 4.5% polyacrylamide gel. The samples

were run on an ABI Prism377 DNA Analyzer.

Page 42: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 41 -

Table 2.4: Microsatellite markers used for genescan analyses shown with the accession number,

fluorescent labelling at the 5’ end of the forward primer, and annealing temperature of the microsatellite,

as well as size range of the PCR product (1Fredholm et al., 1993; 2Winterø & Fredholm, 1995; 3Davies et

al., 1994; 4Rohrer et al., 1994; 5Alexander et al., 1996; 6Joller et al., 2009; 7Karlskov-Mortensen et al.,

2007).

Microsatellite

marker

Accession

no.

Modification at

the 5’ end

Annealing

temperature (°C) Size range (bp)

1S0068 M97244 TET 62 211-260

2S0075 AF044970 FAM/HEX 62 134-162

3S0283 X79925 FAM 62 132-148

4SW207 AF235238 FAM 58 170-188

4SW698 AF235339 TET/FAM 58 194-224

5SW1876 AF253726 FAM 65 204-258

6HSA125gt FM877810 HEX 59 200-240

6MUC4gt FM877809 FAM 59 210-250

7KVL1293 EF131094 HEX 55 249-253

8HEG1T4A --- HEX 53 258-257

8 Microsatellite HEG1T4A (TTTTA repeats) was discovered while sequencing candidate gene HEG1 and

was later registered in dbSNP (NCBI). This microsatellite mapped twice on SSC13, due to an error in the

reference sequence 10.2 at 144695707/144920548 bp.

2.3.5 Pyrosequencing

Pyrosequencing relies on the detection of pyrophosphate release following nucleotide

incorporation (Ronaghi et al., 1998). The analyses were performed using a PyroMark Q24 (Qiagen) at the

Genetic Diversity Centre (GDC), ETH Zurich. To make the templates for pyrosequencing, a single-strand

DNA (ssDNA) is immobilised with the use of streptavidin-coated magnetic beads that bind to the

biotinylated primer incorporated in the sequence, as described in Section 2.3.2.4.

The ssDNA template is hybridised with a sequencing primer and incubated with DNA

polymerase, ATP sulfurylase, luciferase, apyrase, adenosine 5´ phosphosulfate (APS), and luciferin. The

addition of a dNTP starts the sequencing process. The DNA polymerase incorporates the dNTPs into the

template. Pyrophosphate (PPi) is released and then converted in the presence of adenosine 5´

phosphosulfate in ATP by enzyme ATP sulfurylase. The ATP is used by luciferase to convert luciferin

Page 43: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 42 -

into oxyluciferin, generating visible light in an amount proportional to the ATP generated. The light is

detected by a camera and converted to sequence data by software. Unincorporated nucleotides and ATP

are degraded by the apyrase, and the reaction restarts with another nucleotide, until the ssDNA sequence

is determined (Figure 2.2).

Figure 2.2: Example of a pyrogram showing the nucleotide sequence in a specific section of DNA. The

tops represent light emission and nucleotide binding; the height of each peak is proportional to the

number of nucleotides incorporated. SNPs are in the yellow band. A heterozygous SNP has half the signal

strength of a homozygous SNP.

2.3.6 SNP chip

The Porcine SNP60 DNA BeadChip (Illumina, Inc., San Diego, CA, USA) possesses 62,163

probes. It offers comprehensive coverage of the porcine genome for whole-genome association studies,

estimation of genetic breeding value, identification of quantitative trait loci, and comparative genetic

studies (Gunderson et al., 2005; Steemers & Gunderson, 2007; Ramos et al., 2009). Each chip contains 12

independent arrays and can be used to determine genetic variation in porcine breeds such as Duroc,

Landrace, Piétrain, and Large White. The BeadChip was developed through Illumina's iSelect program, in

collaboration with leading porcine researchers from both US and international universities and research

institutes, including Iowa State University, the University of Illinois, Cambridge University, and

Wageningen University. All the SNPs have been deposited in dbSNP (NCBI) under accession numbers

ss131027063 to ss131629651 (Table 2.5).

For our project, we organized a three-generation study on 32 pigs selected from UEH. A blood

sample was taken from all the pigs, after which the DNA was extracted, purified with a GenElute

Mammalian Genomic DNA Miniprep Kit (Sigma-Aldrich), and the concentration measured with the

Page 44: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 43 -

Qubit (Invitrogen), as described in Section 2.3.1. The DNA was then used for the SNP arrays at the

NCCR "Frontiers in Genetics", University of Geneva.

Table 2.5: Selected SNPs belonging to the Illumina Porcine SNP60 BeadChip (Illumina, Inc., San Diego,

CA, USA) (Ramos et al., 2009). Primers are given with the SNPs, DNA fragment size, annealing

temperature, and position of SNPs according to the reference sequence Sscrofa 10.2.

SNPs name Primers Forward and Reverse

Annealing

temperature

(°C)

Amplicon

length

Position

on Sscrofa

10.2

MARC0067282 MARC-1F: TGGGTTAAGGATCCAGCACT

MARC-1R: GTCAAGGACAGGCCAACATT 55 490 144611608

ALGA0072075 ALGA-5F: ATCACCTCCTGGAACCACAG

ALGA-5R: AAAGCTGCGGACAGTGAGAT 57 692 144832256

ALGA0122555 ALGA-1F: CCTTTGACCTACCTGCCTGT

ALGA-1R: GGAGGGTTCCTTTTTCTCCA 54 489 144946317

ALGA0106330 ALGA-4F: CGATCAAGTTCAAGATCTCTTCTG

ALGA-4R: TGACTGTCACATCTTCCTTATCTT 55 249 145009805

ALGA0106230 ALGA-3F: CTGAGACCTGCTGCTGTCC

ALGA-3R: ATCTCCCCCAGCTTCACAAT 55 273 145023374

H3GA0037348 H3GA-1F: CTGAGCTGATGGGGAAGAAC

H3GA-1R: GATCTCAGGCTGATCCCAGA 55 497 ---1

DIAS0000584 DIAS-1AF: TCGGTAAAAGAGTGAGCCTTG

DIAS-1AR: ACTGGGCATGCTGACGTT 56 244 145414267

H3GA0037371 H3GA-2F: CCAGCCATCTCGATTTGC

H3GA-2R: CAGAGCTGATGGGAGAAAGG 55 334 145473321

MARC0006918 MARC-4F: ATTCGGCAATGCCTCTCC

MARC-4R: CCTGCTCACACAGCTTCC 55 400 ---1

MARC0045417 MARC-5F: CAAGAGGGGTGATGCAGAAT

MARC-5R: CTGGTTTTTGTTTGGGGATG 53 396 146679892

ALGA0072168 ALGA-7F: GGCTGAGTACATATCATCCATCC

ALGA-7R: CAATGTAACATCCAAATCTGTTTTT 54 397 147489986

ALGA0072286 ALGA-10F: TGAGTGTTTGATCCAGCTTCC

ALGA-10R: CCAAGACGATACTTTGGATTCA 55 449 153425171

ALGA0072308 ALGA-11F: TTCTGGGGGTGGATTATCAG

ALGA-11R: CATCCCATACCTGTCAGCCTA 55 376 154103780

1 SNPs were not found in reference sequence Sscrofa 10.2.

2.3.7 PCR restriction

The PCR products were digested in a total volume of 25 µl, using 1─2 U restriction enzyme at

an appropriate temperature for 2─16 h, according to the manufacturer’s instructions. Restriction samples

Page 45: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 44 -

were run on agarose gels containing EtBr, to visualise and identify the restriction fragment length

polymorphisms (RFLPs) (Table 2.6).

Table 2.6: Selected SNPs in TNK2, MUC4, HEG1, and MUC13 genes and in the Illumina Porcine SNP60

BeadChip (Illumina, Inc., San Diego, CA, USA) (Ramos et al., 2009), detected by PCR-RFLP.

Restriction enzymes and lengths of the digested fragments are given.

Gene Primers SNP Restriction enzyme Length of digested fragments (bp)

Position of SNPs

TNK2

g.7075 TNK2e6-7-Fc2/R A

TaqI 189, 436

C 625

g.7717 TNK2e9j-F/R T

BseDI 1, 9, 19, 24, 43, 45, 88, 142, 181

C 1, 9, 19, 24, 43, 45, 73, 88, 108, 142

g.11142 TNK2e12b-Fv2/Rv2 A

AluI 18, 27, 97, 134, 214, 261

G 17, 18, 27, 97, 117, 214, 261

MUC4

g.6242 MUC4-6138-F/6887-R A

DdeI 106, 102, 261, 280

G 106, 261, 382

g.7947 MUC4-7741-F/8202-R G

Hin1II 4, 38, 42, 147, 231

A 4, 38, 42, 102, 129, 147

g.8227 MUC4-8012-F/8378-R G

XbaI 155, 212

C 367

HEG1

g.5244 HEG1-F/R T

TspRI 34, 79, 240

C 26, 34, 79, 214

MUC13

226 MUC13-F/R A

FOKI 247, 289

G 33, 214, 289

c.1930 MUC13-ED2-F/R T

HaeIII 61, 242, 393

G 61, 65, 177, 393

MARC0067282 MARC-1F/R T

DdeI 240, 250

C 45, 195, 250

ALGA0122555 ALGA-1F/R T

TaqI 489

C 208, 281

H3GA0037348 H3GA-1F/R A

TaqI 99, 398

C 497

DIAS0000584 DIAS-1AF/R C

PstI 244

G 101, 143

H3GA0037371 H3GA-2F/R A

DdeI 153, 181

G 334

MARC0006918 MARC-4F/R A

DdeI 51, 349

G 51, 168, 181

Page 46: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 45 -

Continued from previous page

Position of SNPs Primers SNP Restriction enzyme Length of digested fragments (bp)

MARC0045417 MARC-5F/R G

TaqI 176, 220

A 396

ALGA0072168 ALGA-7F/R G

HaeIII 195, 202

A 397

ALGA0072286 ALGA-10F/R A

TspRI 86, 363

G 86, 96, 267

ALGA0072308 ALGA-11F/R A

Hin1II 90, 134, 152

G 71, 81, 90, 134

2.3.8 High throughput sequencing

2.3.8.1 Illumina HiSeq 2000

DNA samples from two pigs selected from UEH were used as templates to amplify, by long-

range PCR, the intergenic regions HEG1-MUC13 and MUC13-ITGB5 (Tables 2.2.1 and 2.2.2). The long-

range PCR products were purified as described in Section 2.3.9. The DNA concentration was measured

with the Qubit (Invitrogen), as described in Section 2.3.1. The samples were sent to the Functional

Genomics Center Zurich (FGCZ), a joint state-of-the-art facility of the ETH Zurich and the University of

Zurich, to be sequenced by a HiSeq 2000 (Illumina, Inc., San Diego, CA, USA). The library preparation

was conducted according to Illumina protocols (Figure 2.3).

Figure 2.3: Simplified diagram of the Illumina HiSeq 2000 sequencing procedure (Illumina).

Page 47: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 46 -

2.3.8.2 PacBio RS

Long-range PCR products A and B and L8 in HEG1-MUC13 were later resequenced in both pigs

again at FGCZ, using a PacBio RS (Pacific Biosciences of California, Inc., USA).

The long-range PCR samples were first sheared into a random library of ~1000 bp fragments and

converted into SMRTbell™ library (Travers et al., 2010). The SMRTbell™ structure consists of a

double-stranded portion containing the fragment of interest and a single-stranded hairpin loop on either

end. The hairpin loop provides a site for the primer binding. Sequencing primers were annealed to the

DNA, and a modified DNA polymerase was added in order to form a template-polymerase complex. This

complex was loaded onto a chip containing 150,000 microscopic cavities (zero-mode waveguides or

ZMVs). One complex was immobilised at the bottom of one ZMW. By adding the four differently

labelled nucleotides, the sequencing reaction was started. The fluorescent dye was attached to the

phosphate chain of the nucleotide. The dye was cleaved off with the phosphate chain when the nucleotide

was incorporated by the DNA polymerase and the incorporation of the nucleotide registered base per

base. The cleaved fluorescent dye was detected at the bottom of each chamber; when the dye molecules

started to diffuse away, no fluorescence signal was detected until the incorporation of the next nucleotide

(Figure 2.4).

Figure 2.4: Simplified diagram of the PacBio RS sequencing procedure (Pacific Biosciences).

Page 48: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 47 -

2.3.9 PCR purification

The PCR products for sequencing were purified and concentrated using Montage PCR

centrifugal filter devices (Millipore) or Microcon centrifugal filter units (Millipore), as described in the

manufacturer’s protocol. The PCR products were applied to the filter and supplemented with ddH2O to

400 µl. After centrifugation at 1000 g for 15 min, 21 µl ddH2O were added to the filter. After 5 min at

room temperature, the filter was placed upright on an empty tube and centrifuged at 1000 g for 2 min.

A modified method, described by Boyle & Lew (1995), was also used to purify PCR products

for sequencing and long-range PCRs. The volumes of the PCR products were estimated and 2 vol of

guanidine HCl 6 M were added, then 20 µl of silica suspension were added (Appendix). The DNA was

left binding to the silica beads for 10 min at room temperature; the tubes were inverted several times in

order to keep the beads in suspension. After centrifugation for 1 min at 13,000 rpm, the supernatant was

discarded and the silica pellet was washed with 2 vol of cold wash buffer (Appendix). The solution was

centrifuged again at 13,000 rpm for 1 min, and the supernatant was discarded. The washing step was

repeated twice, and the pellet was left to air dry at room temperature for 10 min. The DNA was

resuspended from the silica beads with 20 µl of distilled water at 55°C. The solution was centrifuged

again at 13,000 rpm for 1 min, and the supernatant containing the DNA was collected in a separate tube.

The DNA concentration was measured with the Qubit (Invitrogen), as described in Section 2.3.1.

A 15 ng/100 bp fragment was sent for sequencing (Microsynth, Balgach).

Long-range PCRs of the intergenic regions HEG1-MUC13 and MUC13-ITGB5 were purified by

using a QIAquick® gel extraction kit (Qiagen). The long-range PCR products were run on 0.8% agarose

gels in 0.5 X TBE buffer containing 100 µg/l EtBr at 75─100 mA for 2 h with a 1 Kb size standard.

Under UV lights, with the use of a scalpel, the expected bands were excised and placed in 1.5 ml tubes.

The gel pieces (100─200 mg) containing the DNA fragments were suspended in 300 µl of Buffer QG

from the kit, incubated at 50°C for 10 min, and vortexed. After the incubation, 100 µl of isopropanol were

added. The solution was transferred to a QIAquick spin column and centrifuged at 13,000 rpm for 1 min.

The flow-through was discarded, and 500 µl of Buffer QG were again added to the column. After

centrifugation, the flow-through was again discarded. The column was washed with 750 µl of Buffer PE

Page 49: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 48 -

and centrifuged for 1 min at 13,000 rpm. The column was placed in a clean 1.5 ml tube, and the DNA

eluted in 30 µl of Buffer EB (10 mM Tris-HCl, pH 8.5) after centrifugation for 1 min at 13,000 rpm.

The DNA concentration was measured with the Qubit (Invitrogen), as described in Section 2.3.1.

The templates were diluted to the concentration required and sent for sequencing to the FGCZ, as

described in Section 2.3.8.

2.4 RNA methods

2.4.1 RNA extraction

Total RNA from intestinal scrapings was extracted with TRIzol (Invitrogen), and the tissue was

deep-frozen in liquid nitrogen. Of the material, 100 mg were homogenised in 1 ml of TRIzol with a

stator-rotor tissue homogeniser. The homogeniser was cleaned before and after each sample with

deionized water, NaOH 1 M, to prevent carry-over of RNA from one sample to another. The homogeniser

was again rinsed in water, dried, and dipped in TRIzol before the homogenisation of the next sample.

The solution was then incubated at room temperature for 5 min, after which 200 µl of chloroform

were added. The sample was vortexed for 15 s and incubated at room temperature for 2─3 min, and then

the solution was centrifuged at 12000 g for 15 min at 4°C. The mixture was separated into a lower red

phase, an interphase containing the genomic DNA, and an aqueous upper phase containing the RNA

(60% of the mixture volume). The aqueous phase was transferred into a new tube, and the RNA was

precipitated by adding 500 µl of isopropyl alcohol. The sample was incubated at room temperature for 10

min, and then centrifuged at 12,000 g for 10 min at 4°C. The RNA formed a gel-like pellet on the sides

and bottom of the tube. The supernatant was removed using a vacuum device, and the pellet was washed

with 1 ml of EtOH 75% and centrifuged at 12,000 g for 5 min at 4°C. The ethanol was removed with the

vacuum device, and the pellet was left to air dry for 5─10 min then resuspended in 30 µl of RNAse-free

water. The sample was incubated for 10 min at 55°C, and stored at -70°C.

Page 50: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 49 -

2.4.2 RNA quantification

RNA concentration was measured using a Qubit® (1.0) fluorometer with a Qubit® Quant-

iTTMRNA assay kit (Invitrogen) or using a NanoDrop ND-1000 spectrophotometer (Thermo Scientific).

Total RNA quality was tested using a 2100 Bioanalyzer, and an RNA 6000 Nano Chip (Agilent

Technologies), provided by the FGCZ. The chips are made of glass and have an interconnected network

of etched micro-channels filled with a gel-dye mixture. Each chip contains 16 wells: three for loading the

gel-dye mixture, one for a molecular size ladder also used as a reference for calculating the concentration

of RNA fragments, and 12 for the samples. The movement of the RNA through the micro-channels is

controlled by a series of electrodes and a software program that allows automated data analysis (Panaro et

al., 2000; Mueller et al., 2000). All the RNA chips were prepared according to the manufacturer’s

instructions, with the materials provided by the kit. After the run, the Bioanalyzer displayed the data as

both migration-time plots and as computer-generated virtual pictures.

Figure 2.5: Electropherograms of total RNA extracted from intestinal scrapings. Left: good quality RNA.

The two main peaks correspond to rRNA subunits 18S and 28S. Right: partially degraded RNA.

2.4.3 Reverse Transcription PCR

The M-MLV reverse transcription system (Promega) was used for reverse transcription-PCR

(RT-PCR) of total RNA. A total of 20 µg of RNA was treated with 30 U of DNase (Qiagen), 10 X of

DNase buffer, and RNase-free water up to 100 µl reaction volume. The samples were incubated at 37°C

for 1 h, and then at 70°C for 5 min to inactivate the enzyme. The RNA was then stored at -70°C or used

for the M-MLV RT-PCR.

The RT-PCR was usually performed with 2 µg of DNase-treated RNA. The RNA was placed in

microcentrifuge tubes with 1 µg of either random nonamers or oligo(dT)17 primers. The samples were

incubated for 5 min at 70°C and then placed on ice. Then, a master mix was added to the sample, for a

Page 51: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 50 -

total volume of 25 µl. The master mix was made with 5 X Reaction Buffer (250 mM Tris-HCl, 375 mM

KCl, 15 mM MgCl2, 50 mM DTT), 5 mM MgCl2, 20 U recombinant RNasin, and 200 U M-MLV reverse

transcriptase. The sample was then incubated for 1 h at 42°C, in cases when oligo(dT)17 primers were

added, or the incubation was at 37°C when random nonamers were used.

To inactivate the M-MLV enzyme, the cDNA was incubated at 95°C for 5 min. The cDNA was

then stored at -20°C or used immediately in a PCR for beta-actin (ACTB) to test the quality of the RT-

PCR. The primers for ACTB were designed from exon sequences. The expected band is around 250 bp if

genomic DNA is used as template and 150 bp with cDNA (Table 2.1). This control PCR shows, under

UV lights after the gel run, if there are possible genomic DNA contaminations in the cDNA samples.

2.4.4 PCR and sequencing

The cDNA was used for specific PCRs with primers designed from exon sequences (Table 2.1).

Usually, 1 µl of cDNA was taken as a template. PCR was performed on the cDNA templates in 25 µl

reaction volume analogous to standard PCR, as described in Section 2.3.2. Purification of the cDNA

PCRs was performed with Microcon centrifugal filter units, as described in Section 2.3.6. The RNA

concentration was measured with the Qubit (Invitrogen), as described in Section 2.3.1. A 15 ng/100 bp

fragment was sent for sequencing (Microsynth, Balgach).

2.4.5 High throughput sequencing

Total RNA from intestinal scrapings was extracted from five pigs selected from UEH. The RNA

was extracted with TRIzol (Invitrogen), as described in Section 2.4.1. RNA concentration was measured

with the Qubit (Invitrogen) or using a NanoDrop (Thermo Scientific), as described in Section 2.4.2. Total

RNA quality was tested using a 2100 Bioanalyzer (Agilent Technologies) at the FGCZ, as described in

Section 2.4.2. The samples were sent to the FGCZ to be sequenced by the SOLiDTM 3 System (Life

Technologies, Inc., Carlsbad, CA, USA). For constructing a library, 500 ng of total RNA depleted of

ribosomal RNA were needed from each sample. The RNA was ligated with adaptors and reverse

transcripted in cDNA. The cDNA was amplified and again size-selected, and the library was ready for

sequencing on the SOLiDTM 3 System.

Page 52: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 51 -

Figure 2.6: Simplified diagram of the SOLiDTM 3 System RNA sequencing procedure (Life

Technologies)

2.5 Computational methods

2.5.1 Linkage analysis

PCR sequences containing SNPs were compared using either Chromas Pro v. 1.5 or CLC

Sequence Viewer v. 6.4 software. The alleles of the microsatellites were analysed with the ABI PRISM®

GeneScan® Analysis Software v. 2.1 software. IGV v. 2.1 software was used to compare the high-

throughput sequencing of the cDNA samples and of the long-range PCR in the intergenic regions HEG1-

MUC13 and MUC13-ITGB5. The haplotypes were determined using Merlin (Abecasis et al., 2002) and

HaploPainter software (Thiele & Nürnberg, 2005).

2.5.2 Statistics of F4ad adhesion

Bacterial adhesion to the enterocytes was either 0% or close to 100% in most UEH pigs tested

with the F4ad MAT, particularly in sites A and D (Section 3.8.1). The percentage of ETEC F4ad adhesive

Page 53: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 52 -

enterocytes is not normally distributed, and a normal distribution could not be achieved even with the

transformation of the data. The data was analysed using the Kruskal-Wallis one-way analysis of variance.

The Kruskal-Wallis is based on ranks and does not require a normal distribution of the data. Statistical

analyses were performed with SYSTAT v. 13 software (Systat SoftwareGmbH, Erkrath, Germany).

Models of inheritance for ETEC F4ad receptor were created by analysing the matings with the

Pedigree Analysis Package v. 4.0 (Department of Human Genetics, University of Utah, UT, USA).

2.5.3 In silico mapping

Human sequences were used for BLAST searches and compared to the pig genome generated by

the Swine Genome Sequencing Consortium (Schook et al., 2005; http://www.sanger.ac.uk/cgi-

bin/blast/submitblast/s_scrofa). Human and pig sequences were compared with the ones present in NCBI

(http://blast.ncbi.nlm.nih.gov/). The physical positions of SNPs and microsatellites were determined by

blasting the sequences in the Pig Genome Database, Sscrofa 10.2 (NAGRP Blast Center;

http://www.animalgenome.org/blast/). The physical positions of the BAC clones to which the sequences

belonged were determined from the BAC fingerprint contig map (Humphray et al., 2007;

http://pre.ensembl.org/Sus_scrofa_map/Info/Index).

Page 54: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 53 -

3. RESULTS

3.1 Exclusion of gene MUC4 as locus for F4bcR

From the SPS, 78 pigs of 38 litters were phenotyped for ETEC F4ab/F4ac susceptibility using

MAT, and their genotype in MUC4_g.8227 was determined by PCR-RFLP (Figure 3.1, Table 2.6). Sixty-

four pigs (82%) were typed as susceptible to ETEC F4ab/F4ac. SNPs MUC4_g.8227 was in LD with the

F4bcR in only 92.3% of the pigs. Pig 2055NO, susceptible to ETEC F4ab/F4ac in the MAT, was

genotyped in MUC4_g.8227 as C/C, the C allele being associated with the absence of ETEC F4ab/F4ac

adhesion. Five pigs (3887C2, 4626II, 6134BLW, 6240PU and 6245PU) were phenotyped as resistant,

whereas their genotype was C/G in MUC4_g.8227 indicating a susceptibility to ETEC F4ab/F4ac.

From the UEH, boar 2349, which was used for breeding purposes, was found to have a

recombination in the MUC4-F4bcR interval. Boar 2349 was homozygous for the susceptible allele at the

F4bcR locus, but heterozygous in SNP MUC4_g.8227. Boar 2349 was mated with four sows that were

resistant to ETEC F4ab/F4ac, generating 48 offspring. The offspring were phenotyped with MAT and

genotyped in MUC4_g.8227 (Figure 3.1). All offspring were susceptible to ETEC F4ab/F4ac; 25 (52.8%)

showed in MUC4_g.8227 as a C/C genotype that is usually associated with the absence of ETEC

F4ab/F4ac adhesion. MAT confirmed the homozygosity for the allele resistant to ETEC F4ab/F4ac in the

sows.

Boar 2349 and its offspring were tested with PCR-RFLP for other SNPs in MUC4. SNPs

MUC4_g.6242 A>G and MUC4_g.7947 G>A are also associated with ETEC F4ab/F4ac

susceptibility/resistance (Figure 3.1, Table 2.6). The other two SNPs were selected according to haplotype

information, based on 10 Nordic Experimental Herd (NEH) pigs and 10 Swiss pigs provided by

Jørgensen’s Danish group. Boar 2349 was heterozygous in both SNPs as in MUC4_g.8227. The offspring

with a C/C genotype in MUC4_g.8227 had a G/G and an A/A genotype in MUC4_g.6242 and

MUC4_g.7947, respectively, associated with the absence of ETEC F4ab/F4ac adhesion.

The parents of 2349, boar 1978 and sow 2002, were genotyped for the same SNPs. Two siblings,

2344 and 2348, from the same litter as 2349, were also genotyped (Figure 3.2, Table 3.1). The pigs were

sequenced in the interval MUC4_g.6138-6887 that contains, together with SNP MUC4_g.6242, another

Page 55: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 54 -

11 SNPs not in LD with F4bcR (Table 3.1). Haplotype analyses showed that 2349 inherited the

recombinant allele in MUC4 from 1978, by a crossing-over event during meiosis (Figure 3.2). The

recombinant allele was then transmitted from 2349 to half its progeny.

Figure 3.1:

Left: Restriction pattern of DdeI digestion of MUC4-6138-F/6887-R 749 bp PCR product to determine

the MUC4_g.6242 A>G polymorphism. Middle: Restriction pattern of Hin1II digestion of MUC4-7741-

F/8202-R PCR products to determine the MUC4_g.7947 G>A polymorphism. Right: Restriction pattern

of XbaI digestion of MUC4-8012-F/8378-R 367 bp PCR product to determine the MUC4_g.8227 G>C

polymorphism.

Page 56: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 55 -

Page 57: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 56 -

Figure 3.2: Diagram of three-generation family tree of 2349 and haplotypes in the SSC13q41-q44 region.

Marker names and position, according to Sscrofa assembly 9, are given on the right side. Animal identity

is shown above the haplotypes. The SNP genotypes are depicted on the left and right sides of the coloured

columns. Digit 1 corresponds to nucleotide A, 2 to C, 3 to G and 4 to T. Microsatellite markers are

represented by 5, 6 and so on, depending on the size of the allelic bands. S above the colored bars

indicates the adhesive haplotype, and s indicates the nonadhesive haplotype. Each haplotype has its own

colour. The lines indicate the relationships among the pigs. The geometric figures above the animal’s

identity indicate the sex: squares indicate males and circles indicate females. The filled figures indicate

ETEC F4ab/F4ac susceptible, and the blank figures represent resistant pigs (Rampoldi et al., 2011

[modified version]).

3.2 Exclusion of interval ZDHHC19-LMLN as locus for F4bcR

For further investigation of the recombinant event in 2349, three SNPs in TNK2 were typed by

PCR-RFLP (Figure 3.3, Table 2.6): TNK2_g.7075 A>C, TNK2_g.7717 T>C and TNK2_g.11142 A>G.

Two SNPs in ZDHHC19 (ZDHHC19_g.4043 T>C) and KIAA0226 (KIAA0226_g.62250 A>G) were

typed by pyrosequencing (Table 2.3). These SNPs were associated with ETEC F4ab/F4ac

susceptibility/resistance by Jørgensen’s Danish group by haplotyping 10 NEH and 10 Swiss pigs. A SNP

in Leishmanolysin-like peptidase (LMLN), LMLN_g.15920, was detected in this study by blasting the

human sequence (NM_001136409) to the pig genome and by sequencing the genomic porcine homolog.

The remaining SNPs in MUC13 and leucine-rich repeat and calponin homology domain-containing

protein 3 (LRCH3) were selected according to the literature.

Haplotype analyses showed that 2349 still possessed a recombinant allele in interval ZDHHC19-

LMLN, but not in MUC13 (Figure 3.2). Boar 2349’s recombination ended in the interval between genes

LMLN-MUC13 (~808 Kb). Two SNPs tested in MUC13, MUC13-226 and MUC13-813, were in LD with

F4bcR in 2349’s family (Table 3.1).

SNP MUC13-226 is mapped in an intronic region of MUC13, and it is a restriction site for

enzyme FOKI (Figure 3.4, Table 2.6). SNP MUC13-813 is mapped in an exon. However, the C>T

transition is a silent mutation. SNP MUC13-813 was tested in our study by pyrosequencing (Table 2.3).

Page 58: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 57 -

Table 3.1: Polymorphisms in genes ZDHHC19, TNK2, MUC4, KIAA0226, LRCH3, LMLN and MUC13

of 2349’s family, which were determined by sequencing, pyrosequencing, and PCR-RFLP. SNPs name

and changes in nucleotides are given. The ETEC F4ab/F4ac phenotype is given as S for susceptibility and

s for resistance.

F4bcR S>s S/S S/S s/s S/s S/s

SNPs 2349 2348 2344 1978 2002

ZDHHC19

g.4043 T>C T/C T/T C/C T/C T/C

TNK2

g.7075 A>C A/C A/A C/C A/C A/C

g.7717 T>C T/C T/T C/C T/C T/C

g.11142 A>G A/G A/A G/G A/G A/G

MUC4

g.6242 G>A G/A A/A G/G G/A G/A

g.6308 G>T G/T T/T G/G G/T G/T

g.6317 G>A G/A A/A G/G G/A G/A

g.6321 G>C G/C C/C G/G G/C G/C

g.6609 T>A A/A A/A T/A A/A A/T

g.6616 G>T T/T T/T G/T T/T T/G

g.6634 A>C C/C C/C A/C C/C C/A

g.6675 Del GAACGT/No Del Del Del Hetero Del Hetero

g.6690 A>T T/T T/T A/T T/T A/T

g.6745 T>C C/C C/C T/C C/C C/T

g.6770 G>T T/T T/T G/T T/T T/G

g.6862 T>C C/C C/C T/C C/C C/T

g.7947 A>G A/G A/A G/G A/G A/G

g.8227 G>C G/C G/G C/C G/C G/C

KIAA0226

g.62250 A>G A/G A/A G/G A/G A/G

LRCH3

212 G>C G/C G/G G/C G/C G/G

213 G>C G/C G/G G/C G/C G/G

214 T>C T/C T/T T/C T/C T/T

215 T>A T/A T/T T/A T/A T/T

216 No Del/Del A Hetero No Del Hetero Hetero No Del

217 G>A G/A G/G G/A G/A G/G

218 A>G A/G A/A A/G A/G A/A

219 G>T G/T G/G G/T G/T G/G

LMLN

g.15920 G>C G/C G/G G/C G/C G/G

MUC13

227 T>C T/T T/T T/C T/C T/T

226 A>G A/A A/A G/G A/G A/G

225 C>G C/C C/C C/G C/G C/C

224 G>A G/G G/G G/A G/G G/A

Page 59: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 58 -

Continued from previous page

F4bcR S>s S/S S/S s/s S/s S/s

SNPs 2349 2348 2344 1978 2002

MUC13

223 C>T C/C C/C C/T C/C C/T

813 C>T C/C C/C T/T C/T C/T

814 G>T G/G G/G T/T G/G G/T

829 A>C A/A A/A C/C A/A A/C

895 A>C A/A A/A C/C A/A A/C

905 G>A G/G G/G A/A G/G G/A

908 G>A G/G G/G A/A G/G G/A

920 A>G A/A A/A G/G A/A A/G

933 C>T C/C C/C T/T C/C C/T

935 A>C A/A A/A A/A A/A A/A

Figure 3.3:

Left: Restriction pattern of TaqI digestion of TNK2e6-7-Fc2/R PCR product to determine the

TNK2_g.7075 A>C polymorphism. Middle: Restriction pattern of BseDI digestion of TNK2e9j-F/R PCR

product to determine the TNK2_g.7717 T>C polymorphism. Right: Restriction pattern of AluI digestion

of TNK2e12b-Fv2/Rv2 PCR product to determine the TNK2_g.11142 A>G polymorphism.

Page 60: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 59 -

Figure 3.4:

Left: Restriction pattern of TspRI digestion of HEG1-F/R PCR product to determine the HEG1_g.5244

T>C polymorphism. Middle: Restriction pattern of FOKI digestion of MUC13-F/R PCR product to

determine the MUC13-226 A>G polymorphism. Right: Restriction pattern of HaeIII digestion of

MUC13-ED2-F/R PCR product to determine the MUC13_c1930 T>G polymorphism.

3.3 SNPs chip results

To investigate further the recombinant event in 2349, we performed a three-generation study

using the Porcine SNP60 DNA BeadChip. Thirty-two pigs were selected, including 2349, its parents, two

siblings, and 18 offspring from four different matings. Prof. Martien Groenen and his group at Animal

Breeding and Genetics (Wageningen University, Netherlands) provided the SNPs sequence and their

position in the reference sequence 9.0.

The BeadChip contained 62163 SNPs. Of these, only 3638 were located on SSC13. The

recombinant haplotype of boar 2349 started ~6.4 Mb distal to gene ZDHHC19. Sixty-three SNPs were

located in the interval ZDHHC19-S0075 (Table 3.2).

Four SNPs derived by the BeadChip were in complete LD with F4bcR in boar 2349’s family

(Figure 3.2, Table 3.2): SNP ALGA0072075 is mapped in the intergenic region SLC12A8-HEG1, SNP

ALGA0106330 is mapped in an intron of gene MUC13, SNP DIAS0000584 is mapped in an intron of

gene Kalirin (KALRN), and SNP MARC0006918 is mapped in an intron of gene MYLK. Pig 2349’s

recombination ended at SNP ALGA0072075 (Figure 3.2). The SNP was mapped to reference sequence

10.2 at 144832256 bp in SSC13.

Page 61: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 60 -

Table 3.2: List of SNPs from the Porcine SNP60 DNA BeadChip located in the interval ZDHHC19-

S0075. Genes and microsatellites are written in bold. The position of SNPs, microsatellites and the mean

position of genes are according to reference sequence 10.2. SNPs in LD with F4bcR are highlighted.

ZDHHC19 143310000 ALGA0106230 145023374

H3GA0037318 143315618 ITGB5 145060000

ALGA0072055 143574087 S0283 145083113

H3GA0037333 143593394 H3GA0037348 ---2

MARC0012378 143618378 H3GA0037351 ---2

M1GA0017682 143624457 ALGA0072090 145096895

MARC0093203 143638483 MARC0096736 145146697

TNK2 143650000 DIAS0001297 145176833

ASGA0058885 143656188 ALGA0072091 ---2

MUC4 143810000 MARC0112804 145223359

ASGA0058906 143825858 ALGA0072095 145300325

ALGA0072062 143866440 ALGA0115627 145338186

KIAA0226 143890000 ALGA0072104 145547495

ALGA0072071 144065180 KALRN 145410000

ASGA0058918 144082720 DIAS0000584 145414267

MARC0089106 144094647 MARC0021405 145431613

LRCH3 144100000 H3GA0037371 145473321

MARC0043596 144126389 ALGA0072105 145504531

ALGA0072067 144145817 ALGA0072101 145578358

ALGA0072065 144167475 ALGA0072097 145598524

ALGA0072072 144197577 ASGA0058947 145611904

LMLN 144200000 ASGA0058958 145732401

MARC0096777 144499692 MARC0088848 145772058

MARC0099692 144488410 ALGA0105068 146120125

ZNF148 144540000 ASGA0096469 146122112

MARC0067282 144611608 MYLK 146200000

SLC12A8 144850000 MARC0006918 ---2

ALGA0072075 1448322561 ASGA0058970 ---2

HEG1 144800001 H3GA0037376 146288325

MARC0002946 1448101001 H3GA0037373 146265419

ASGA0058923 1447818091 ASGA0058962 146244907

INRA0041036 1447600371 ALGA0072134 146374488

ASGA0058925 1447330311 ALGA0072138 146403234

MARC0006663 1447023921 ALGA0072128 146534002

ALGA0122555 144946317 H3GA0037388 146433577

ASGA0089965 144946742 ASGA0058976 146458592

ASGA0091537 144981309 ASGA0058980 146496697

MUC13 144990000 MARC0053131 146460232/1466045633

ALGA0106330 145009805 S0075 146639048 1 Inversion in Sscrofa 10.2 2 SNP does not map on Sscrofa 10.2 3 SNP maps twice on Sscrofa 10.2

All the SNPs are enlisted according to their physical positions in the genomic DNA

Page 62: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 61 -

The results of the Porcine SNP60 DNA BeadChip and the genotyping of ZDHHC19-MUC13

interval indicated that 2349’s recombination ended between SNPs LMLN_g.15920-ALGA0072075, a ~0.6

Mb interval comprising genes zinc finger protein 148 (ZNF148) and SLC12A8.

No informative SNPs derived by the BeadChip were found in the LMLN_g.15920-

ALGA0072075 interval.

3.4 Exclusion of interval LMLN-ZNF148 as locus for F4bcR

A second three-generation study on 2349 was perfomed to refine the position of F4bcR locus in

SSC13. More markers were genotyped in the ZNF148-ITGB5 interval (Figure 3.5).

The markers were either selected by literature or detected in this study by BLASTing the human

sequences to the pig genome and sequencing the genomic porcine homolog (Table 2.1).

SNP HEG1_g.5244 was tested by PCR-RFLP (Figure 3.4).

Page 63: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 62 -

Page 64: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 63 -

Figure 3.5: Diagram of a second three-generation family tree of 2349 and haplotypes in the SSC13q41-

q44 region. Marker names are given on the left side. Animal identity is shown above the haplotypes. The

SNP genotypes are depicted on the left and right sides of the colored columns. Digit 1 corresponds to

nucleotide A, 2 to C, 3 to G, and 4 to T; 998 corresponds to nucleotide insertion; and 999 corresponds to

nucleotide deletion. Microsatellite markers KVL1293 and HEG1T4A are represented by numbers

corresponding to their peaks in the genescan. S above the coloured bars indicates the adhesive haplotype,

and s indicates the nonadhesive haplotype. Each haplotype has its own colour. The lines indicate the

relationships among the pigs. The geometric figures above animal identity indicate the sex: squares

indicate males and circles indicate females. The filled figures indicate ETEC F4ab/F4ac susceptible, and

the blank figures represent resistant pigs.

Haplotype analyses showed that 2349’s recombination ended in SNP SLC12A8_g.22157, located

in intron 1 of SLC12A8. The SNP was mapped at 144631142bp on reference sequence 10.2. The

sequencing of 2349 excluded that the locus for F4bcR is located in the ZDHHC19-ZNF148 interval and

suggested it may be located in the candidate genes SLC12A8, HEG1, MUC13, or ITGB5.

SNPs in the interval ITGB5_c.246-MARC0006918 were not in LD with F4bcR in boar 2240

(Figure 3.5). SNP SLC12A8_159 was not in LD with F4bcR in all pigs (Figure 3.5). Microsatellite

KVL1293 was in LD with F4bcR in boar 2349’s family. This microsatellite was mapped in intron 12 of

HEG1.

3.5 Exclusion of interval SLC12A8-KVL1293 as locus for F4bcR

Boar 2349 was no longer informative in refining the F4bcR locus in the SLC12A8-ITGB5

interval. Six pigs from SPS with a recombination in MUC4-F4bcR (Section 3.1) were genotyped in the

MUC4-MARC0045417 interval to obtain new information on F4bcR locus position (Table 3.3).

SNPs DIAS0000584 and MARC0006918 were not in LD with F4bcR in the SPS pigs (Table 3.3).

In pigs 3887C2, 4626II, and 6134BLW, the recombination ended proximal to SNP

ZNF148_g.96828. Pigs 6240PU and 6245PU possessed recombinant alleles until SNP SLC12A8_c.2947.

Pig 2055NO possessed a recombinant allele until microsatellite KVL1293, with the exception of SNP

SLC12A8_c.1990 (Table 3.3).

Page 65: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 64 -

SLC12A8_c.1990 T>C transition encoded for a silent mutation in SLC12A8. This SNP is in LD

with F4bcR in 2055NO, but not in 6240PU and 6245PU. In the interval between SLC12A8_g.129502-

SLC12A8_c.1990-SLC12A8_c.2947, no SNPs in LD with F4bcR were discovered.

Table 3.3: Haplotypes of the six pigs recombinant in MUC4-F4bcR from the SPS showing exclusion of

the MUC4-KVL1293 interval as a locus for F4bcR. SNPs name and nucleotide changes are given.

Microsatellite and peaks in genescan are given. The ETEC F4ab/F4ac phenotype is given as S for

susceptibility and s for resistance.

F4bcR S>s s/s s/s s/s s/s s/s S/s

SNPs 6134BLW 4626II 3887C2 6240PU 6245PU 2055NO

MUC4

g.8227 G>C G/C G/C G/C G/C G/C C/C

ZNF148

g.96828 T>G G/G G/G G/G T/G T/G G/G

SLC12A8

g.129502 T>C C/C C/C C/C T/C T/C C/C

c.1990 G>A A/A A/A A/A G/A G/A G/A1

c.2947 T>A A/A A/A A/A A/T A/T A/A

HEG1-MUC13

ALGA0072075 T>C C/C C/C C/C C/C C/C C/C

HEG1

g.5244 T>C C/C C/C C/C C/C C/C C/C

c.321 T>C C/C C/C C/C C/C C/C C/C

g.49379 T>C C/C C/C C/C C/C C/C C/C

KVL1293 253>249/261 249/249 249/249 249/249 249/249 249/249 249/249

MUC13

g.15376 A>G G/G G/G G/G G/G G/G A/G

ALGA0106330 G>A A/A A/A A/A A/A A/A G/A

226 A>G G/G G/G G/G G/G G/G A/G

813 T>C C/C C/C C/C C/C C/C T/C

DIAS0000584 C>G G/G G/G C/G C/G G/G C/G

MARC0045417 A>G A/G G/G G/G A/G G/G G/G 1 Typing error: unfortunately, pig 2055NO could not be retyped.

The interval between markers KVL1293-HEG1T4A-JN613413-23529 (~28 Kb) was sequenced

in unrelated pigs by Jørgensen’s Danish group. The region was highly conserved with an average of 1

SNP every 2000-2500 bp. All the markers discovered were not in LD with F4bcR. SNP JN613413-23529

is located ~2 Kb after HEG1 3’UTR (Table 2.1).

Page 66: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 65 -

3.6 Partial exclusion of MUC13 as locus for F4bcR

Sow 9480XJZ had a recombination in the F4bcR-MUC13 interval (Figure 3.6). Sow 9480XJZ

was resistant in MAT to ETEC F4ab/F4ac and had a C/C genotype in MUC4_g.8227 associated with the

absence of ETEC F4ab/F4ac adhesion. Sow 9480XJZ possessed a heterozygous genotype in MUC13

SNPs: ALGA0106330, MUC13-226, and MUC13-813.

Sow 9480XJZ was mated with four boars resistant to ETEC F4ab/F4ac, generating 21 offspring.

Boar 552, used for mating, was also an offspring of 9480XJZ (Figure 3.6). The offspring were

phenotyped with the MAT, and their genotype in MUC4_g.8227 and MUC13-226 was determined by

PCR-RFLP (Figures 3.1 and 3.4). The offspring were resistant to ETEC F4ab/F4ac and showed a C/C

genotype in MUC4_g.8227.

The PCR-RFLP in MUC13-226 (Figure 3.4) showed that 18 offspring (85.7%) had the same

heterozygous genotype in MUC13-226 as 9480XJZ. Unfortunately, none of the pigs with the recombinant

allele was selected as a breeder. 9480XJZ was genotyped in the MUC4-MARC0045417 interval together

with three of its offspring from two litters (Figures 3.6).

Page 67: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 66 -

Figure 3.6: Family tree and haplotypes of sow 9480XJZ in the MUC4-MARC0045417 region. Marker

names are given on the left side. Animal identity is shown above the haplotypes. The nonadhesive

haplotype is indicated by s. The lines indicate the relationships among the pigs. The colour of the elliptic

circles indicates the SNPs and the microsatellite alleles: orange represents those associated with

susceptibility to F4bcR and green represents resistance. The underlined markers are the ones with high

LD with F4bcR. The red rectangle showed the interval where the pigs are recombinant.

Page 68: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 67 -

Sow 9480XJZ had a possible recombinant event in F4bcR-MUC13 interval. In the MUC4-

KVL1293 interval, 9480XJZ possessed a haplotype associated with resistance to ETEC F4ab/F4ac in

markers with high LD with F4bcR (Figure 3.6). The same associated haplotype was inherited in most of

its offspring.

Sow 9480XJZ’s family was genotyped in gene MUC13, from the 5’UTR until the 3’UTR. It was

not possible to sequence exon 2 of MUC13 because of the presence of tandem repeats. Sow 9480XJZ and

its recombinant offspring possessed a heterozygous or homozygous genotype associated with

susceptibility to ETEC F4ab/F4ac in all the markers of MUC13.

It was not possible to determine by haplotype analyses if the recombination of sow 9480XJZ

started before or after the exon 2 of MUC13 (MUC13_g.1974-MUC13_g.8951). In the MUC13_g.207-

MUC13_g.1974 and MUC13_g.8951-MUC13_g.15150 interval, sow 9480XJZ possessed a homozygous

genotype associated with susceptibility to ETEC F4ab/F4ac. SNP MUC13_g.15376 was mapped in intron

5 of MUC13. Haplotype analyses showed that 9480XJZ’s recombination ended after SNP

JN613413_115242, mapped in the first ~8 Kb of the intergenic region MUC13-ITGB5 (Table 2.1, Figure

3.6). The interval MUC13_g.15150-JN613413_115242 is excluded as a locus for F4bcR.

3.7 Sequencing and mRNA expression of candidate genes

As described in section 2.4.5, the total RNA from intestinal scrapings was extracted from five

pigs and used for high throughput sequencing by the SOLiDTM 3 System. Two pigs were homozygous

susceptible to ETEC F4ab/F4ac (2349 and 641), two were heterozygous (2938 and 2939), and one was

resistant (1168).

The total RNA from intestinal scrapings was also extracted from other five pigs: two were

homozygous susceptible to ETEC F4ab/F4ac (654, 2006), one was heterozygous (2762), and two were

resistant (1017, 1715). The 10 cDNA samples were used to test SNPs in SLC12A8, HEG1, MUC13, and

ITGB5.

Page 69: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 68 -

3.7.1 SLC12A8

The data obtained by the SOLiDTM System displayed a low expression of gene SLC12A8 in the

small intestine. No alternative splicings and no remarkable differences were seen between pigs resistant

and susceptible to ETEC F4ab/F4ac.

Two SNPs, SLC12A8_c.1990 and SLC12A8_c.2947, were found in high LD with F4bcR (Table

2.1). The G>A transition in SLC12A8_c.1990 is a silent mutation, and SNP SLC12A8_c.2947 is located in

the 3’UTR of SLC12A8.

3.7.2 HEG1

The data obtained by the SOLiDTM System displayed a low expression of gene HEG1 in the

small intestine. An alternative splicing was seen in gene HEG1, not related to F4bcR.

The genomic sequence of HEG1 presents an 1147 bp duplication, mapped at 144789345-

144788199 and 144790896-144789750 in reference sequence 10.2. This repeat region contains exon 5 of

HEG1 (351 bp). Primers HEG-E5F/R were designed to amplify a region of 493 in the cDNA of HEG1

between exons 4 and 6 (Table 2.1). After the gel run, a band of ~1000 bp was visible. This band indicates

a duplication of HEG1 in exon 5. Therefore, an alternative form of protein HEG1 is expressed in the

small intestine.

The cDNA was re-extracted from the intestinal tissues, and the PCR was repeated to ensure that

the ~1000 bp band was not an artefact. The cDNA obtained from brain, liver, and muscle tissue were used

in the same PCR. No expression of HEG1 was revealed, except in the intestinal tissue. The PCR products

from two pigs, one homozygous susceptible and the other resistant to ETEC F4ab/F4ac, were sent for

sequencing.

The ~1000 bp band possessed two copies of the exon 5 of HEG1 and a 282 bp alternative exon

was mapped between exons 5 and 6 of HEG1 (Figure 3.7). The duplication and the new exon were in

frame with the mRNA. Four SNPs were found in this cDNA region. However, they were not in LD with

F4bcR. The data obtained by the SOLiDTM displayed low expression in the region where exons 4 and 6

were mapped in the reference sequence 10.2. No RNA transcripts were mapped to the reference sequence

10.2 in the interval corresponding to exon 5, the duplicated exon 5, or the new exon.

Page 70: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 69 -

SNPs were discovered also in exons 3, 6, and 17 of HEG1 but they were also not in LD with F4bcR. Only

SNP HEG1_c.321, mapped in exon 2 of HEG1, was in high LD with F4bcR (Table 2.1). The T>C

transition in HEG1_c.321 is a silent mutation.

Figure 3.7: Alternative splicing of gene HEG1 in exon 5. Primers HEG-E5F/R are highlighted in azure;

SNPs are highlighted in red.

3.7.3 MUC13

The data obtained by the SOLiDTM System displayed high expression of gene MUC13 in the small

intestine. In the interval corresponding to exon 2 of MUC13, RNA transcripts were mapped to the

reference sequence 10.2 only in pigs that were resistant (1168) and heterozygous susceptible (2938 and

2939) to ETEC F4ab/F4ac.

SNP MUC13_c.232 (Table 2.1), mapped in exon 4 of MUC13, generates a stop codon in the

mRNA sequence with a C>T transition. However, the SNP was not in LD with F4bcR (Figure 3.6). SNP

MUC13-813 encodes a silent mutation, as described in section 3.2. SNP MUC13_c.1788 (Table 2.1),

mapped in MUC13 3’UTR, was in high LD with F4bcR, except in 9480XJZ’s recombinant family (Figure

Page 71: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 70 -

3.6). SNP MUC13_c.1930 was tested by PCR-RFLP (Figure 3.4). However, the SNP was not in LD with

F4bcR.

3.7.4 ITGB5

The data obtained by the SOLiDTM System displayed low expression of gene ITGB5 in the small

intestine. No alternative splicings and no remarkable differences were seen between pigs that were

resistant and susceptible to ETEC F4ab/F4ac.

Six SNPs were found in the cDNA of ITGB5: ITGB5_c.246, ITGB5_c.917, ITGB5_c.920,

ITGB5_c.1580, ITGB5_c.1715, and ITGB5_c.2744 (Table 2.1). The six SNPs were not in full LD with

F4bcR (Figure 3.5).

3.8 Sequencing of intergenic regions

3.8.1 Interval of HEG1-MUC13

Long-range PCRs covered the intergenic region HEG1-MUC13 and exon 2 of MUC13 (Table

2.2.1, Figure 3.8). DNA samples from boar 2349 and sow 9480XJZ, respectively homozygous susceptible

and resistant to ETEC F4ab/F4ac, were used as templates.

Long-range PCR products from L1 to L6 were correctly mapped by blast to the reference

sequence 10.2. However, problems in alignment have occurred in long-range PCR products L7, L8 and

L9. The genomic sequences obtained were highly conserved. The alignment with the reference sequence

10.2 has led to the discovery of ~250 SNPs. Sow 9480XJZ possessed a heterozygous or homozygous

genotype associated with susceptibility to ETEC F4ab/F4ac in all but two SNPs, SNP 1 and SNP 2. The

two SNPs were mapped at 144944384 bp (SNP 1 C>A) and at 144978031bp (SNP 2 C>A) (Figure 3.9).

Long-range PCR products A, B and L8 were sequenced by the PacBio platform RS (Table

2.2.1). Long-range PCR products A and B were blasted to the reference sequence 10.2. However,

problems in alignment were seen again in long-range PCR product L8. Sow 9480XJZ possessed a

homozygous genotype associated with susceptibility to ETEC F4ab/F4ac in the sequences derived from

long-range PCR products A and B.

Page 72: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 71 -

Several attempts with different PCR primers were made to amplify exon 2 of MUC13; however,

they were not successful.

3.8.2 Interval of MUC13-ITGB5

Long-range PCRs covered the intergenic region MUC13-ITGB5 and the 5’ of ITGB5 (Table

2.2.2, Figure 3.8). DNA samples from pigs 2349 and 9480XJZ were used as templates.

Long-range PCR products from L1 to L11 were sequenced by the Illumina platform and

correctly mapped by blast to the reference sequence 10.2. The alignment with the reference sequence 10.2

has led to the discovery of ~300 SNPs. Sow 9480XJZ possessed a homozygous genotype associated with

resistance to ETEC F4ab/F4ac in ~200 SNPs. The majority of SNPs were mapped either in the intergenic

region MUC13-ITGB5 or in introns of ITGB5. Three SNPs, ITGB5_c.246, ITGB5_c.917, and

ITGB5_c.920, were mapped in exons of ITGB5 but are not in LD with F4bcR (Figures 3.5 and 3.6).

Figure 3.8: Position of long-range PCR products in the HEG1-ITGB5 interval. The genes, exon order, and

scale are deduced from the reference sequence 10.2.

Page 73: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 72 -

Figure 3.9: SNP positions in the HEG1-ITGB5 interval. The genes, exon order, and scale are deduced

from the reference sequence 10.2. Informative SNPs for pig 9480XJZ are indicated.

Page 74: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 73 -

3.9 Validation of alternative markers for ETEC F4ab/F4ac susceptibility

Analyses of the pigs belonging to UEH and the six pigs recombinant in MUC4-F4bcR from the

SPS have shown that SNPs ALGA0072075, ALGA0106330, MUC13-226, MUC13-813, and microsatellite

KVL1293 were high in LD with F4bcR. The markers in UEH pigs had close to 100% accuracy with the

only exceptions being the pigs in sow 9480XJZ’s recombinant family.

Forty pigs were randomly selected from the SPS as representative of the Swiss porcine

population. A standard MAT was conducted on these pigs and DNA samples were collected. The MAT

revealed that only 13 pigs (32.5%) were resistant to ETEC F4ab/F4ac adhesion. The DNA of the pigs was

initially tested with old marker MUC4_g.8227. Unlike the previous 78 pigs selected from the SPS, which

contained six pigs recombinant in MUC4 gene, the expected genotype of SNP MUC4_g.8227 in the 40

pigs coincided 100% with the phenotype found in the MAT.

The 13 pigs resistant in the MAT were found to have a C/C genotype in MUC4_g.8227 that was

associated with the absence of ETEC F4ab/F4ac adhesion. Among the remaining 27 pigs, eight were

found to be G/G in MUC4_g.8227, which indicated a possible homozygous susceptible genoptype in

F4bcR, whereas the other 19 pigs showed a heterozygous genotype.

The pigs were genotyped in SNPs ALGA0072075, ALGA0106330, MUC13-226, MUC13-813,

and microsatellite KVL1293. SNP 1 and SNP 2, discovered in the intergenic region HEG1-MUC13

(Section 3.8.1), were also tested in the 40 pigs.

Results were similar to SNP MUC4_g.8227. The alleles, associated with susceptibility and

resistance to ETEC F4ab/F4ac, coincided with the phenotypes in all 40 pigs. The markers were in 100%

LD with F4bcR.

The markers could be used in breeding programs to select pigs resistant to F4bcR, as

replacements for SNP MUC4_g.8227. SNP MUC13-813 is currently being patented by Zhang et al.

(2008) for this purpose.

Page 75: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 74 -

Table 3.4: Genotyping of the 461 pigs selected from the SPS in markers MUC4_g.8227, ALGA0072075,

KVL1293, SNP 1, SNP 2, ALGA0106330, MUC13-226, and MUC13-813 compared to their phenotypes

for ETEC F4ab/F4ac susceptibility/resistance.

F4bcR phenotypes

Discordance % on

total Susceptible Resistant

SNPs Genotype SNPs Genotype SNPs

AA AB BB AA AB BB

MUC4_g.8227 8 19 1 5 13 6 13%

ALGA0072075 8 19 1 18 1 2.2%

KVL1293 8 19 1 18 1 2.2%

SNP 1 8 20 18

SNP 2 8 19 12 18 12 2.2%2

ALGA0106330 8 20 18

MUC13-226 8 20 18

MUC13-813 8 20 18 1 Combined data of selected 40 SPS pigs and the 6 SPS pigs recombinant in MUC4_g.8227 (Section 3.5).

2 Typing error: unfortunately, pig 2055NO could not be retyped.

Page 76: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 75 -

3.10 F4ad susceptibility

From the UEH, a total of 489 pigs from sixty-three litters were used to elucidate the inheritance

of the receptors for ETEC F4ad (F4adR). Offspring and parents were divided into three classes based on

the results of the F4ad MAT, as described in section 2.2.4.

The phenotype C (F4ab+/F4ac-/F4ad+) was seen in 26 pigs from the 489 tested with F4ad MAT

(Table 3.5). Most pigs with a C phenotype originated from litters with a common parent. All the C

phenotyped pigs expressed the weak F4ad receptor.

Table 3.5: Phenotypes observed in the 489 pigs from UEH according to the binding of ETEC F4 variants.

The bacterical adhesion is marked with ●.

Phenotypes Fimbrial variants

F4ab F4ac F4ad No. pigs % in the UEH

A ● ● ● 301 61.6

B ● ● 29 5.9

C ● ● 26 5.3

D ● 37 7.6

E 96 19.6

F ● 0 0

G ● 0 0

H ● ● 0 0

Total 489 100

3.10.1 Two receptors for E. coli F4ad

In the offpring and parents, bacterial adhesion to the enterocytes in the four sites examined was

close either to 0%, indicating a resistant phenotype, or to 100%, indicating a fully susceptible phenotype.

In the F4ad MAT, several parents and offspring showed variable percentages of enterocytes susceptible to

ETEC F4ad, especially in sites B and C of the intestine, indicating a third weakly susceptible phenotype.

The weakly susceptible phenotype was not an artefact caused by poor cell quality or mistakes performed

in the F4ad MAT. We found evidence in this study that the weakly susceptible phenotype is inherited

independently from the fully susceptible phenotype, which indicates the existence of at least two

Page 77: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 76 -

receptors in the enterocytes for ETEC F4ad adhesion. We gave the designation E1 to the receptor

responsible for full susceptibility and the designation E2 to the receptor responsible for weak

susceptibility.

A threshold deduced from observations was made to distinguish pigs resistant (R) to ETEC F4ad

from those having fully susceptible (E1) phenotype or a weakly susceptible (E2) phenotype. Pigs with 0%

adhesive enterocytes in all the four intestinal sites were considered resistant. Pigs with an adhesion

between 0.5% and 100% in at least one of the intestinal sites examined were considered E2, and pigs with

>85% adhesion in all the four intestinal sites were considered E1.

In the matings, it was observed that only pigs with 0% in all sites were truly resistant. Pigs with

even <1% of adhesion were shown to produce susceptible progeny if mated with resistant pigs. Based on

this data, 173 pigs (35.4% of total) were classified as E1, 189 (38.6%) as E2, and 127 (26%) as resistant

to ETEC F4ad (Table 3.6).

Table 3.6: E. coli F4ad adhesion strengths (% of enterocytes with more than 5 adherent bacteria) at four

intestinal sites in pigs of phenotypes E1 (n=173), E2 (n=189) and R (n=127).

Site A Site B Site C Site D

Phenotype E1 E2 R E1 E2 R E1 E2 R E1 E2 R

Minimum 95 0 0 90 0 0 90 0 0 90 0 0

Maximum 100 100 0 100 100 0 100 100 0 100 100 0

Mean 99.5 9.8 0 99.3 16.8 0 99.7 44.3 0 99.4 59.9 0

Standard

deviation 1.6 22.8 0 2.1 27.9 0 1.4 36.7 0 2.0 34.5 0

The pigs possessing an E2 phenotype showed large variability in the F4ad MAT. During the first

years, as described in section 2.2.2, the standard MAT caused some E2 pigs to be labelled erroneously as

resistant to ETEC F4ad or E1. The investigation of four sites decreased significantly the possibility of

mistyping. However, E2 pigs can still be typed with a wrong phenotype. Mistakes can occur during the

F4ad MAT because of the manual selection and counting of the intact enterocytes. Testing more intestinal

sites appears more important than the selection and counting of cells, where the same source of error

would apply to E1 and R pigs.

Page 78: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 77 -

3.10.2 Inheritance of phenotype

The 19 parents were divided into the three phenotypes (E1, E2 and R) according to the F4ad

MAT results. The 470 offspring from the 63 litters were divided into six combinations, based on the

parents’ F4adR phenotypes.

The matings of R x R pigs has generated only resistant pigs (Table 3.7). The matings of E1 x E1

pigs has generated only E1 pigs. The matings of R x E2, R x E1, E1 x E2 and E2 x E2 has generated E1,

E2 and resistant pigs (Table 3.7). The results of the F4ad MAT test on the progeny indicated the existence

of a genetic component in the inheritance of E1 and E2 receptors (Table 3.8). The data, however,

excluded that E1 is a Mendelian monogenetic trait. E1 must be encoded by more than one gene to explain

its expression in E2 x E2 and R x E2 litters, as shown in Table 3.7. The mean adhesion strength observed

in Table 3.8 shows similar values for E1 x R and E1 x E2 matings, indicating that the genes involved in

the inheritance of E1 are independent from E2. Statistical analyses of the data were performed to confirm

this hypothesis (Table 3.9).

Table 3.7: Distribution of the three phenotypes for ETEC F4d in progeny with known phenotyped

parents.

Phenotype of parents Number of progeny Phenotype of progeny

E1 E2 R

R x R 45 0 0 45

R x E1 178 81 79 18

R x E2 107 6 55 46

E1 x E1 63 63 0 0

E1 x E2 36 12 15 9

E2 x E2 41 9 28 4

Page 79: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 78 -

Table 3.8: Influence of phenotypes of parents on the median adhesion strengths (± SD) at the four

intestinal sites A through D of the progeny.

Phenotypes

of parents

Number of

matings

Number of

progeny

Mean adhesion strength (% of enterocytes with more than 5

adherent bacteria) ± standard deviation (SD) at intestinal site

A B C D

R x R 7 45 0.0±0.0 0.0±0.0 0.0±0.0 0.0±0.0

R x E1 16 178 49.5±48.4 52.2±46.8 68.6±39.1 74.4±36.4

R x E2 15 107 8.3±25.1 10.7±27.1 20.5±34.2 31.1±38.5

E1 x E1 11 63 99.9±0.6 99.6±1.6 99.8±0.9 99.8±1.1

E1 x E2 5 36 42.7±48.6 43.7±46.5 49.9±47.8 52.1±46.2

E2 x E2 9 41 31.1±40.3 43.9±43.5 54.9±42.2 66.6±37.2

Table 3.9: Kruskal-Wallis test with the six mating combinations of the three phenotypes for ETEC F4ad.

Significant differences (p<0.05) between the combinations are in bold.

Site A

Matings R x R R x E1 R x E2 E1 x E1 E1 x E2 E2 x E2

R x R --- 0.000 0.005 0.000 0.000 0.000

R x E1 --- 0.000 0.000 0.353 0.052

R x E2 --- 0.000 0.000 0.000

E1 x E1 --- 0.000 0.000

E1 x E2 --- 0.566

E2 x E2 ---

Site B

Matings R x R R x E1 R x E2 E1 x E1 E1 x E2 E2 x E2

R x R --- 0.000 0.001 0.000 0.000 0.000

R x E1 --- 0.000 0.000 0.325 0.197

R x E2 --- 0.000 0.000 0.000

E1 x E1 --- 0.000 0.000

E1 x E2 --- 0.921

E2 x E2 ---

Site C

Matings R x R R x E1 R x E2 E1 x E1 E1 x E2 E2 x E2

R x R --- 0.000 0.000 0.000 0.000 0.000

R x E1 --- 0.000 0.000 0.045 0.039

R x E2 --- 0.000 0.001 0.000

E1 x E1 --- 0.000 0.000

E1 x E2 --- 0.650

E2 x E2 ---

Page 80: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 79 -

Continued from previous page

Site D

Matings R x R R x E1 R x E2 E1 x E1 E1 x E2 E2 x E2

R x R --- 0.000 0.000 0.000 0.000 0.000

R x E1 --- 0.000 0.000 0.010 0.101

R x E2 --- 0.000 0.010 0.000

E1 x E1 --- 0.000 0.000

E1 x E2 --- 0.263

E2 x E2 ---

The results of the Kruskal-Wallis tests indicated that E1 and E2 are not encoded by the same

receptor gene. If the same gene expresses receptors E1 and E2, no significant difference should be present

between mating combinations, such as E1 x E1 and E2 x E2 or R x E1 and R x E2. The test showed a

significant difference in all four sites.

E2 pigs have a crescent adhesion strength that can be 0% in sites A and B or >85% in sites C and

D. This crescent adhesion explains why pairs of mating combinations, such as E1 x E2 and R x E1, show

a significant difference only in sites A and B.

3.10.3 Statistical pedigree analysis

Models for the inheritance of F4adR were evaluated with the Pedigree Analysis Package v. 4.0

(Department of Human Genetics, University of Utah, UT, USA) at the Institute of Genetics, University of

Berne.

The phenotype was assigned to three classes: resistant, weakly, or fully susceptible to ETEC

F4ad. Five different models were considered in the analysis. A general genetic model estimating the allele

frequency, the transmission probabilities, the dominance effect, the displacement to characterize a major

gene, and the heritability to characterize a polygenic component was compared with an environmental

model, where a major gene was excluded by setting the transmission probabilities equal to the allele

frequency. In the case where the general genetic model turns out to explain the data better than the

environmental model, it is compared to a mixed inheritance model that is the very same model, with the

exception that the transmission probabilities are set to be Mendelian. In the case where the mixed

inheritance model turns out to be better than the general genetic model it is compared to a major gene

model and a polygenic model. The major gene model is the same as the mixed inheritance model with the

Page 81: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 80 -

exception that a polygenic component is excluded by setting the heritability to zero. The polygenic model

only estimates the heritability.

The data show that E1 and E2 are not the product of a single receptor gene, confirming the

results of the Kruskal-Wallis test (Table 3.9).

The mixed inheritance model explained our data best (Table 3.10). It seems that the ETEC F4ad

receptors are regulated by both a major gene and a polygenic component. This is in accordance with our

proposed model for the inheritance of F4adR in the pig.

We postulated that the E1 phenotype is encoded by two complementary or epistatic genes, with

the alleles A and a, and B and b respectively, while the E2 phenotype is encoded by only one gene with

the alleles W and w (Table 3.11). The receptor E1 is expressed only when both hypothetical alleles, A and

B, occur in the dominant form, either homozygously or heterozygously. The receptor E2 is expressed as a

Mendelian dominant trait. The expression of phenotype E1 covers the expression of phenotype E2.

Several matings with many offspring are necessary to be sure of a breeder genotype in F4adR.

Table 3.10: Evaluation of the Pedigree Analysis Package models. The differences in the -2- ln likelihoods

of the different models follow a chi-square (χ2) distribution where the degrees of freedom (df) are equal to

the difference in the number of parameters estimated. Significant differences (p<0.01) are in bold.

Models df -2- ln likelihood χ2 df p

general genetic

environmental

7

3

727.2

1061.7 334.5 4 <0.001

general genetic

mixed inheritance

7

4

727.2

729.0 1.8 3 0.626

mixed inheritance

major gene

4

3

729.0

767.0 38.0 1 <0.001

mixed inheritance

polygenic

4

1

729.0

757.4 28.4 3 <0.001

The -2- ln likelihood of the mixed inheritance model is not significantly different from the -2- ln

likelihood of the general model. Therefore it is considered the better model as it needs less parameters to

explain the data.

Page 82: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 81 -

Table 3.11: Analyses of the largest litters (≥12 pigs) from UEH. Repetitive matings were put together.

Hypothetical genotypes of parents were deduced from phenotypes observed in progeny. For each litter,

the parents, the number of piglets, the phenotypes for ETEC F4ad, and F4dR hypothetical genotype are

indicated. The genotype frequencies for systems E1 and E2, the expected phenotypes for ETEC F4ad, and

the obtained phenotypes are shown for each litter. Chi-square (χ2) for the Hardy-Weinberg equilibrium is

calculated for each litter. A separate χ2 is calculated only for E1 phenotype. Significant differences

(p<0.05) are in bold.

Litters Parents Offspring

χ2 No. pigs N°

Phen

otype Genotype

System E1 System E2 Expected Obt.

Gen./Frequency % Phen./Freq. %/No. No.

217 & 230 2831

2842

R

R

aa Bb ww

aa bb ww

aa bb

aa Bb

50

50 ww 100

R

E2

E1

100

0

0

24

0

0

24

0

0

0.00 24

205 & 222 2674

2577

E1

R

AA Bb WW

aa Bb ww

Aa bb

Aa B-

25

75 Ww 100

R

E2

E1

0

25

75

0

6.8

20.2

0

9

18

0.95

0.24 E1 27

216 & 2231 2674

2570

E1

R

AA Bb WW

aa bb ww

Aa bb

Aa Bb

50

50 Ww 100

R

E2

E1

0

50

50

0

15.5

15.5

2

11

18

0.81

If R=E2

0.4 E1 31

2272 2674

2840

E1

R

AA Bb WW

aa bb ww

Aa bb

Aa Bb

50

50 Ww 100

R

E2

E1

0

50

50

0

7

7

3

8

3

4.57

If R=E2

2.29 E1 14

2281 2674

2842

E1

R

AA Bb WW

aa bb ww

Aa bb

Aa Bb

50

50 Ww 100

R

E2

E1

0

50

50

0

6.5

6.5

1

6

6

0.08

If R=E2

0.04 E1 13

232 & 244 2831

2791

R

E1

aa Bb ww

Aa BB WW

aa B-

Aa B-

50

50 Ww 100

R

E2

E1

0

50

50

0

6

6

0

9

3

3.00

1.50 E1 12

238 2988

2577

E1

R

AA Bb WW

aa Bb ww

Aa bb

Aa B-

25

75 Ww 100

R

E2

E1

0

25

75

0

3.8

11.2

0

5

10

0.51

0.13 E1 15

248 2859

2842

E1

R

Aa Bb Ww

aa bb ww

aa bb

aa Bb

Aa bb

Aa Bb

25

25

25

25

ww

Ww

50

50

R

E2

E1

37.5

37.5

25

4.9

4.9

3.2

7

3

3

1.65

0.01 E1 13

219 2831

2726

R

E2

aa Bb ww

aa bb Ww

aa bb

aa Bb

50

50

ww

Ww

50

50

R

E2

E1

50

50

0

7

7

0

6

8

0

0.29 14

221 & 231 2831

2766

R

E2

aa Bb ww

Aa bb Ww

aa bb

Aa bb

aa Bb

Aa Bb

25

25

25

25

ww

Ww

50

50

R

E2

E1

37.5

37.5

25

7.5

7.5

5

11

7

2

3.47

1.80 E1 20

Page 83: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 82 -

Continued from previous page

Litters Parents Offspring

χ2 No. pigs N°

Phen

otype Genotype

System E1 System E2 Expected Obt.

Gen./Frequency % Phen./Freq. %/No. No.

229 2554

2726

R

E2

aa bb ww

aa bb Ww aa bb 100

ww

Ww

50

50

R

E2

E1

50

50

0

6

6

0

7

5

0

0.33 12

2341 2831

2878

R

E2

aa Bb ww

aa bb WW

aa bb

aa Bb

50

50 Ww 100

R

E2

E1

0

100

0

0

12

0

2

10

0

0.00

If R=E2 12

213 2732

2791

E1

E1

AA Bb WW

Aa BB WW A- B- 100 WW 100

R

E2

E1

0

0

100

0

0

13

0

0

13

0.00 13

207 2559

2450

E2

E2

Aa bb WW

aa Bb Ww

Aa bb

aa Bb

Aa bb

Aa Bb

25

25

25

25

W- 100

R

E2

E1

0

75

25

0

9

3

0

11

1

1.78

1.33 E1 12

1 In litters 216, 223, 228, and 234, based on the parents supposed genotype, resistant pigs were not

expected. If the pigs were counted as E2, the χ2 showed no significant difference (p<0.05).

2 In litter 227, based on the parents supposed genotype, resistant pigs were not expected. χ2 showed a

significant difference (p<0.05), even if the pigs counted as resistant were calculated as E2.

Unexpected phenotypes in litters could be caused by mistyping in the F4ad MAT. By testing

only four sites in the F4ad MAT, not all E2 pigs could be distinguished from E1 or resistant pigs.

The significative differences observed in litters 227 could be caused by a mistake in assigning the

hypothetical genotype of the parents. A separate χ2 was calculated in litters with expected E1 phenotype

to test the two genes’ inheritance model for the E1 receptor. The data showed no significative difference

in all litters examined for E1.

Page 84: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 83 -

4. DISCUSSION

4.1 F4bcR mapping on SSC13

The genotyping of pigs from the UEH revealed the presence of many haplotypes associated with

resistance to ETEC F4ab/F4ac in the ALGA0072075-MUC13-813 interval (Figures 3.2, 3.5, and 3.6). The

haplotype associated with susceptibility to ETEC F4ab/F4ac in the interval ALGA0072075-MUC13-813

was identical in all the pigs genotyped. The presence of several haplotypes associated with resistance to

ETEC F4ab/F4ac indicated that the pig was not originally susceptible to ETEC F4ab/F4ac. A mutation in

the DNA provokes the expression of a receptor on the brush border of the enterocytes, enabling the

adhesion of ETEC F4ab/F4ac. Because of the presence of different haplotypes, until the causative

mutation is discovered all markers used for the selection of pigs resistant to F4bcR will not be 100%

accurate.

The recombinant allele in 2349 allowed the exclusion of the whole ZDHHC19-MUC4-ZNF148

interval as a position for the F4bcR locus (Section 3.4, Figure 3.5). Boar 2240, resistant to ETEC

F4ab/F4ac in MAT, possessed a homozygous genotype associated with susceptibility to ETEC F4ab/F4ac

in all the SNPs tested downstream ITGB5. It is possible that 2240 had a recombination in the interval

ITGB5-MYLK similar to the one in ZDHHC19-ZNF148 of pig 2349 (Section 3.4, Figure 3.5).

The data from pig 2349 indicated that the F4bcR locus should be mapped to one of the four

candidate genes: SLC12A8, HEG1, MUC13, and ITGB5.

4.2 Exclusion of genes SLC12A8 and HEG1

The RNA sequencing results from the SOLiDTM System revealed a low level of expression for

genes SLC12A8 and HEG1 in the small intestine. No RNA expression was revealed in the intronic

sequences of the genes or in the intergenic region SLC12A8-HEG1. Exon 5 duplication seen in HEG1 was

not associated with ETEC F4ab/F4ac susceptibility. A similar duplication was not present in the human

homolog gene (ENSG00000173706). No expression of HEG1 was revealed in brain, liver, and muscle, so

it is not clear whether exon duplication leads to an alternative splicing expressed only in the intestine.

Page 85: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 84 -

More analyses are necessary in other tissues where the gene is expressed more strongly. SNP

SLC12A8_159, reported to be in LD with F4bcR by Huang et al. (2008), showed no association with

ETEC F4ab/F4ac susceptibility in the second three-generation study conducted on pig 2349 (Figure 3.5).

Pig 2055NO from SPS showed a recombinant haplotype in all the SNPs tested in the interval SLC12A8-

HEG1, with the exception of SLC12A8_c.1990 (Table 3.3). Two other pigs from the SPS, 6240PU and

6245PU, showed a recombinant genotype in SLC12A8_c.1990 that encodes for a silent mutation. The

interval between SLC12A8_c.1990-SLC12A8_c.2947 was sequenced in unrelated pigs. All the markers

discovered were not in LD with F4bcR, indicating a typing error in pig 2055NO that unfortunately could

not be retyped.

Microsatellite KVL1293 was mapped to intron 12 of HEG1. The interval between KVL1293 and

the 3’UTR of HEG1 was sequenced in unrelated pigs by Jørgensen’s Danish group. All the markers

discovered were not in LD with F4bcR. The data obtained from SPS pigs disprove the genome-wide

association study by Fu et al. (2012), which demonstrated an association between HEG1 and F4bcR. Our

results suggested that the F4bcR locus does not map in the region SLC12A8-HEG1.

4.3 Exclusion of genes ITGB5

The data from the SOLiDTM System revealed a low level of expression for genes ITGB5 in the

small intestine. No RNA expression was revealed in the intronic sequences of the gene or in the

intergenic region MUC13-ITGB5. No SNPs in LD with F4bcR were found in the exonic sequences of

ITGB5 in UEH pigs (Figures 3.5 and 3.6). The data disprove Huang et al. (2011), who reported SNPs

ITGB5_c.920, ITGB5_c.1580, ITGB5_c.1715, and ITGB5_c.2744 in LD with F4bcR.

The comparison of all the results showed that it is highly probable that ITGB5 is not the locus of

F4bcR.

4.4 MUC13

No remarkable differences in expression were observed in MUC13 between intestinal samples

from pigs resistant or susceptible to ETEC F4ab/F4ac, confirming the findings of Schroyen et al. (2012).

Page 86: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 85 -

No RNA expression was revealed in the intronic sequences of MUC13 or in the intergenic region HEG1-

MUC13.

Recombinant sow 9480XJZ, resistant to ETEC F4ab/F4ac in the MAT, was sequenced in the

whole gene MUC13, except for the exon 2 (MUC13_g.1974-MUC13_g.8951). In all the SNPs tested,

from 5’UTR to 3’UTR, either a heterozygous or a homozygous genotype associated with susceptibility to

ETEC F4ab/F4ac was observed (Figure 3.6).

RNA reads from ETEC F4ab/F4ac homozygous susceptible pigs 641 and 2349 could not be

mapped in exon 2 of MUC13 (Section 3.8.1). Exon 2 of MUC13 is a 3-5 kb region rich in tandem repeats.

Ren et al. (2012) found differences in the tandem repeat sequences between pigs susceptible and resistant

to ETEC F4ab/F4ac (Section 1.8.2.2). The reference sequence 10.2 is based on the genomic DNA of a

Duroc pig resistant to ETEC F4ab/F4ac. Only the RNA reads from pigs with a resistant allele to ETEC

F4ab/F4ac could be mapped to the reference sequence 10.2.

The data indicated that the tandem repeat region in the exon 2 of MUC13 is the most probable

location for the F4bcR locus.

4.5 F4adR inheritance

The F4ad MAT and the statistical analyses revealed the presence of a weakly susceptible

receptor for ETEC F4ad (E2), as postulated by Hu et al. (1993). The E2 receptor, however, was found to

be expressed in breeder pigs from the UEH. The hypothesis that expression of the weak receptor for F4ad

is terminated at 16 weeks of age is not valid; like the receptor E1, the receptor E2 is expressed throughout

the lifespan.

In most of the E2 pigs, an increase was observed in the adhesion strength of the bacteria the

closer the intestinal sites were to the ileocaecal valve. Inhibitors of gene expression could explain the

inhomogeneous presence of the weak ETEC F4ad receptor in the small intestine.

Our model for F4adR inheritance is mostly valid and confirms Bijlsma & Bouw’s (1987)

asssumption of the existence of more than one gene controlling the F4ad receptor. Few litters did not

respect the Hardy-Weinberg equilibrium with unexpected phenotypes (Table 3.11). The unexpected

phenotypes could be caused by mistakes in the F4ad MAT or in typing the hypothetical genotypes of the

parents in F4adR. Mutation in the receptors for ETEC F4ad could also be involved.

Page 87: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 86 -

In the past years, several pigs from the UEH were reported expressing a C phenotype

(F4ab+/F4ac-/F4ad+) in the MAT (Python, 2003; Python et al., 2005). Most pigs with a C phenotype

originated from litters with a common parent, indicating that a genetic component may be involved in

phenotype expression. ETEC F4ab adhesion was tested in the MAT by examining only one intestinal site.

This test could have led some pigs used in this study, with a possible C phenotype, to be labelled as D

(F4ab-/F4ac-/F4ad+). All the C phenotyped pigs expressed the weak F4ad receptor. It is possible that like

ETEC F4ad, ETEC F4ab adhesion is also governed by two receptors: F4bcR and another one involved

with the C phenotype. It is also possible that ETEC F4ab can weakly bind to a mutated ETEC F4ad E2

receptor. More data are necessary to investigate this hypothesis.

4.6 Conclusions and perspectives

The different tandem repeat sequences observed by Ren et al. (2012) are the most probable cause

of ETEC F4ab/F4ac susceptibility in pigs. Pig 9480XJZ could express in MUC13 either the variant void

of the O-glycosylated binding site or a third variant with a shorter glycosylation site because of its

recombination. Bacteria could not use a shorter glycosylation site as a binding site. If further attempts at

DNA and cDNA sequencing on the tandem repeats prove futile, proteomic analyses from intestinal

scrapings of 9480XJZ’s recombinant progeny could determine which variant is expressed in MUC13.

The sequencing problem of MUC13 exon 2 precludes its use as a valuable marker. Because of

the possibility of close cross-over events, future diagnostic tests for F4ab/F4ac susceptibility should be

performed using at least two markers close to both ends of exon 2. The markers described in section 3.9

have proven quite reliable, whereas the two newly discovered SNPs of the intergenic region HEG1-

MUC13 need more analyses to ensure their complete LD with the F4bcR (Section 3.8.1 and Figure 3.9).

Pigs should be tested for E2 phenotype at more than four intestinal sites to show in detail the

expression of the weak receptor in the intestine. If further matings prove that the E2 phenotype is

inherited as a dominant monogenetic trait, genome scans will be performed in selected pigs to find

possible candidate genes for the receptor locus.

Page 88: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 87 -

REFERENCES:

Abecasis G.R., Cherny S.S., Cookson W.O., Cardon L.R. (2002): Merlin-rapid analysis of dense genetic

maps using sparse gene flow trees. Nature Genetics 30: 97-101.

Alexa P., Štouračová K., Hamřík J., Rychlík I. (2001): Gene typing of the colonisation factors K88 (F4)

in the enterotoxigenic Escherichia coli strains isolated from diarrhoeic piglets. Veterinární Medicína 46:

46-49.

Alexander L.J., Rohrer G.A., Beattie C.W. (1996): Cloning and characterization of 414 polymorphic

porcine microsatellites. Animal Genetics 27: 137-148.

Atroshi F., Alaviuhkola T., Schildt R., Sandholm M. (1983): Fat globule membrane of sow milk as a

target for adhesion of K88-positive Escherichia coli. Comparative Immunology, Microbiology and

Infectious Diseases 6: 235-245.

Baker D.R., Billey L.O., Francis D.H. (1997): Distribution of K88 Escherichia coli adhesive and

nonadhesive phenotypes among pigs of four breeds. Veterinary Microbiology 54: 123-132.

Bentley R. & Meganathan R. (1982): Biosynthesis of vitamin K (menaquinone) in bacteria.

Microbiological Reviews 46: 241-280.

Berberov E.M., Zhou Y., Francis D.H., Scott M.A., Kachman S.D., Moxley R.A. (2004): Relative

importance of heat-labile enterotoxin in the causation of severe diarrheal disease in the gnotobiotic piglet

model by a strain of enterotoxigenic Escherichia coli that produces multiple enterotoxins. Infection and

Immunity 72: 3914-3924.

Bijlsma I.G.W., De Nijs A., Van Der Meer C., Frik J.F. (1982): Different pig phenotypes affect adherence

of Escherichia coli to jejunal brush-borders by K88ab, K88ac, or K88ad antigen. Infection and Immunity

37: 891-894.

Bertschinger H.U., Moon H.W., Whipp S.H. (1972): Association of Escherichia coli with the small

intestinal epithelium. Infection and Immunity 5: 595-605.

Bijlsma I.G.W. & Bouw J. (1987): Inheritance of K88-mediated adhesion of Escherichia coli to jejunal

brush borders in pigs: A genetic-analysis. Veterinary Research Communications 11: 509-518.

Billey L.O., Erickson A.K., Francis D.H. (1998): Multiple receptors on porcine intestinal epithelial cells

for the three variants of Escherichia coli K88 fimbrial adhesin. Veterinary Microbiology 59: 203-212.

Page 89: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 88 -

Björkstén B., Sepp E., Julge K., Voor T., Mikelsaar M. (2001): Allergy development and the intestinal

microflora during the first year of life. Journal of Allergy and Clinical Immunology 108: 516-520.

Blomberg L., Henriksson A., Conway P.L. (1993): Inhibition of adhesion of Escherichia coli K88 to

piglet ileal mucus by Lactobacillus spp. Applied and Environmental Microbiology 59: 34-39.

Bork P., Downing A.K., Kieffer B., Campbell I.D. (1996): Structure and distribution of modules in

extracellular proteins. Quarterly Reviews of Biophysics 29: 119-167.

Bosi P., Casini L., Finamore A., Cremokolini C., Merialdi G., Trevisi P., Nobili F., Mengheri E. (2004):

Spray-dried plasma improves growth performance and reduces inflammatory status of weaned pigs

challenged with enterotoxigenic Escherichia coli K88. Journal of Animal Science 82: 1764-1772.

Boyle J.S. & Lew A.M. (1995): An inexpensive alternative to glassmilk for DNA purification. Trends in

Genetics 11: 8.

Brown E.A. & Hardwidge P.R. (2007): Biochemical characterization of the enterotoxigenic Escherichia

coli LeoA protein. Microbiology 153: 3776-3784.

Camerlink I., Ellinger L., Bakker E.J., Lantinga E.A. (2010): Homeopathy as replacement to antibiotics in

the case of Escherichia coli diarrhea in neonatal piglets. Homeopathy 99: 57-62.

Castillo M., Martìn-Orùe S.M., Taylor-Pickard J.A., Pérez J.F., Gasa J. (2008): Use of

mannanoligosaccharides and zinc chelate as growth promoters and diarrhea preventative in weaning pigs:

Effects on microbiota and gut function. Journal of Animal Science 86: 94-101.

Chandler D.S., Mynott T.L., Luke R.K.J., Craven J.A. (1994): The distribution and stability of

Escherichia coli K88 receptor in the gastrointestinal tract of the pig. Veterinary Microbiology 38: 203-

215.

Chappuis J.P., Duval-Iflah Y., Ollivier L., Legault C. (1984): Escherichia coli K88 adhesion: A

comparison of Chinese and Large White piglets. Genetic Selection Evolution 16: 385-390.

Chen X., Gao S., Jiao X., Liu X.F. (2004a): Prevalence of serogroups and virulence factors of

Escherichia coli strains isolated from pigs with postweaning diarrhoea in eastern China. Veterinary

Microbiology 103: 13-20.

Chen X.Q., Zhang X.D., Liang R.Q., Cao M.Q. (2004b): Betaine improves LA-PCR amplification.

Chinese Journal of Biotechnology 20: 715-718.

Page 90: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 89 -

Clarke S.C. (2001): Diarrhoeagenic Escherichia coli-an emerging problem? Diagnostic Microbiology &

Infectious Disease 41: 93-98.

Cox E. & Houvenaghel A. (1993): Comparison of the in vitro adhesion of K88, K99, F41 and P987

positive Escherichia coli to intestinal villi of 4- to 5-week-old pigs. Veterinary Microbiology 34: 7-18.

Curtis J. & Bourne F.J. (1971): Immunoglobulin quantitation in sow serum, colostrum and milk and the

serum of young pigs. Biochimica et Biophysica Acta 236: 319-332.

Davies W., Høyheim B., Chaput B., Archibald A.L., Frelat G. (1994): Characterization of microsatellites

from flow-sorted porcine chromosome 13. Mammalian Genome 5: 707-711.

De Geus B., Harmsen M., Van Zijderveld F. (1998): Prevention of diarrhoea using pathogen specific

monoclonal antibodies in an experimental enterotoxigenic E. coli infection in germfree piglets. The

Veterinary Quarterly 20: S87-S89.

De Greve H., Wyns L., Bouckaert J. (2007): Combining sites of bacterial fimbriae. Current Opinion in

Structural Biology 17: 506-512.

Delsol A.A., Randall L., Cooles S., Woodward M.J., Sunderland J., Roe J.M. (2005): Effect of the growth

promoter avilamycin on emergence and persistence of antimicrobial resistance in enteric bacteria in the

pig. Journal of Applied Microbiology 98:564-571.

Downing A.K., Knott V., Werner J.M., Cardy C.M., Campbell I.D., Handford P.A. (1996): Solution

structure of a pair of calcium-binding epidermal growth factor-like domains: implications for the Marfan

syndrome and other genetic disorders. Cell 85: 597-605.

Duchet-Suchaux M.F., Bertin A.M., Menanteau P.S. (1991): Susceptibility of Chinese Meishan and

European Large White pigs to enterotoxigenic Escherichia coli strains bearing colonization factor K88,

987P, K99, or F41. American Journal of Veterinary Research 52: 40-44.

Dunn D.L. (1990): Autochthonous microflora of the gastrointestinal tract. Perspectives on Colon Rectal

Surgery 2: 105-119.

ECH, European Committee of Homeopathy, Spence D., Nicolai T., Van Wassenhoven M., Ives G.

(2005): A strategy for research in homeopathy: assessing the value of homeopathy for health care in

Europe. 3rd edition: 1-27.

Eckburg P.B., Bik E.M., Bernstein C.N., Purdom E., Dethlefsen L. Sargent M., Gill S.R., Nelson K.E.,

Relman D.A. (2005): Diversity of the human intestinal microbial flora. Science 308: 1635-1638.

Page 91: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 90 -

Edfors-Lilja I., Petersson H., Gahne B. (1986): Performance of pigs with or without the intestinal receptor

for Escherichia coli K88. Animal Production 42: 381-387.

Edfors-Lilja I., Gustafsson U., Duval-Iflah Y., Ellergren H., Johansson M., Juneja R.K., Marklund L.,

Andersson L. (1995): The porcine intestinal receptor for Escherichia coli K88ab, K88ac: regional

localization on chromosome 13 and influence of IgG response to the K88 antigen. Animal Genetics 26:

237-242.

EFSA, European Food Safety Authority (2007): Foodborne antimicrobial resistance as a biological

hazard-Scientific Opinion of the Panel on Biological Hazards. Question No. EFSA-Q-2007-089

http://www.efsa.europa.eu/en/efsajournal/doc/765.pdf

Elton C.M., Smethurst P.A., Eggleton P., Farndale R.W. (2002): Physical and functional interaction

between cell-surface calreticulin and the collagen receptors integrin α2β1 and glycoprotein VI in human

platelets. Thrombosis and Haemostasis 88: 648-654.

Engel P. (1998): Genetische Untersuchungen über Rezeptoren für Escherichia coli F4 (K88) im Darm

von Schweinen als Grundlage für züchterische Massnahmen zur Bekämpfung der Diarrhöe bei Ferkeln.

Dissertation, Universität Giessen.

Engel P., Seibert B., Beuing R, Senft B., Erhardt G. (1998): Occurence of Escherichia coli F4 receptors in

several pig breeds in Germany and influence on parameters of reproduction and fattening performance.

Archives of Animal Breeding 41: 75-87.

Fairbrother J.M. (1999): Identification, nomenclature and diagnosis of pathogenic Escherichia coli.

Proceedings of the Annual Meeting of the Western Canadian Association of Swine Practitioners.

Saskatoon, Canada: 21-31.

Fairbrother J.M., Nadeau E., Gyles C.L. (2005): Escherichia coli in postweaning diarrhea in pigs: An

update on bacterical types, pathogenesis and prevention strategies. Animal Health Research Reviews 6:

17-39.

Fairbrother J.M. & Gyles C.L. (2006): Escherichia coli infections. In: Taylor D.J.: Disease of Swine. 9th

edition, Wiley-Blackwell, Oxford, UK: 639-674.

Floss D.M., Falkenburg D., Conrad U. (2007): Production of vaccines and therapeutic antibodies for

veterinary applications in transgenic plants: an overview. Transgenic Research 16: 315-332.

Fredholm M., Winterø A.K., Christensen K., Kristensen B., Nielsen P.B., Davies W., Archibald A.

(1993): Characterization of 24 porcine (dA-dC)n-(dT-dG)n microsatellites: genotyping of unrelated

animals from four breeds and linkage studies. Mammalian Genome 4: 187-192.

Page 92: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 91 -

Frenyó V.L., Pethes G., Antal T., Szabó I. (1981): Changes in colostral and serum IgG content in swine in

relation to time. Veterinary Research Communications 4: 275-282.

Frydendahl K. (2002): Prevalence of serogroups and virulence genes in Escherichia coli associated with

postweaning diarrhoea and edema disease in pigs and a comparison of diagnostic approaches. Veterinary

Microbiology 85: 169-182.

Fu W.X., Liu Y., Lu X., Niu X.Y., Ding X.D., Liu J.F., Zhang Q. (2012): A Genome-Wide Association

Study Identifies Two Novel Promising Candidate Genes Affecting Escherichia coli F4ab/F4ac

Susceptibility in Swine. PloS One 7: e32127.

Fürer E., Cryz S.J. Jr., Dorner F., Nicolet J., Wanner M., Germanier R. (1982): Protection against

colibacillosis in neonatal piglets by immunization of dams with procholeragenoid. Infection and

Immunity 35: 887-894.

Galindo-Rodriguez S.A., Allemann E., Fessi H., Doelker E. (2005): Polymeric nanoparticles for oral

delivery of drugs and vaccines: a critical evaluation of in vivo studies. Critical Reviews in Therapeutic

Drug Carrier Systems 22: 419-464.

Glenn G.M., Flyer D.C., Ellingsworth L.R., Frech S.A., Frerichs D.M., Seid R.C., Yu J. (2007):

Transcutaneous immunization with heat-labile enterotoxin: development of a needle-free vaccine patch.

Expert Review of Vaccines 6: 809-819.

Gross L. (2006): Bacterial Fimbriae Designed to Stay with the Flow. PLoS Biology 4: e314.

Guarner F. & Malagelada J.R. (2003): Gut flora in health and disease. Lancet 361: 512-519.

Guérin G., Duval-Iflah Y., Bonneau M., Bertaud M., Guillaume P., Ollivier L. (1993): Evidence for

linkage between K88ab, K88ac intestinal receptors to Escherichia coli and transferrin loci in pigs. Animal

Genetics 24: 393-396.

Guinée P.A. & Jansen W.H. (1979): Behaviour of Escherichia coli K antigens K88ab, K88ac, and K88ad

in immunoelectrophoresis, double diffusion, and hemagglutination. Infection and Immunity 23: 700-705.

Gunderson K.L., Steemers F.J., Lee G., Mendoza L.G., Chee M.S. (2005): A genome-wide scalable SNP

genotyping assay using microarray technology. Nature Genetics 37: 549-554.

Gyles C.L. (1994): Escherichia coli verotoxin and other cytotoxin. In: Gyles C.L.: Escherichia coli in

domestic animals and humans. CAB International, Wallingford, UK: 151-170.

Page 93: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 92 -

Hediger M.A., Romero M.F., Peng J.B., Rolfs A., Takanaga H., Bruford E.A. (2004): The ABCs of solute

carriers: physiological, pathological and therapeutic implications of human membrane transport proteins:

Introduction. European Journal of Physiology 447: 465-468.

Hewett D., Samuelsson L., Polding J., Enlund F., Smart D., Cantone K., See C.G., Chadha S., Inerot A.,

Enerback C., Montgomery D., Christodolou C., Robinson P., Matthews P., Plumpton M., Wahlstrom J.,

Swanbeck G., Martinsson T., Roses A., Riley J., Purvis I. (2002): Identification of a psoriasis

susceptibility candidate gene by linkage disequilibrium mapping with a localized single nucleotide

polymorphism map. Genomics 79: 305-314.

Holt J.P., Van Heugten E., Graves A.K., See M.T., Morrow W.E.M. (2011): Growth performance and

antibiotic tolerance patterns of nursery and finishing pigs fed growth-promoting levels of antibiotics.

Livestock Science 136: 184-191.

Hu Z.L., Hasler-Rapacz J., Huang S.C., Rapacz J. (1993): Studies in swine on inheritance and variation in

expression of small intestinal receptors mediating adhesion of the K88 enteropathogenic Escherichia coli

variants. The Journal of Heredity 84: 157-165.

Huang L., Ren J., Yan X., Huang X., Zhang Z., Zhang B. (2006): MUC13 molecular marker for

identifying the F4ac adhesion-caused diarrhea resistance of the weaning pig and the use thereof. Patent

number WO2010066118.

Huang X., Ren J., Yan X., Peng Q., Tang H., Zhang B., Ji H., Yang S., Huang L. (2008): Polymorphisms

of three gene-derived STS on pig chromosome 13q41 are associated with susceptibility to enterotoxigenic

Escherichia coli F4ab/ac in pigs. Science in China. Series C: Life Sciences 51: 614-619.

Huang L., Ren J., Yan X.M. (2011): cDNA cloning of porcine ITGB5 gene and its association with

susceptibility to ETEC F4ac. http://www.agrpaper.com; epub ahead of print.

Hudault S., Guignot J., Servin A.L. (2001): Escherichia coli strains colonizing the gastrointestinal tract

protect germ-free mice against Salmonella typhimurium infection. Gut 49: 47-55.

Hüffmeier U., Lascorz J., Traupe H., Böhm B., Schürmeier-Horst F., Ständer M., Kelsch R., Baumann C.,

Küster W., Burkhardt H., Reis A. (2005): Systematic linkage disequilibrium analysis of SLC12A8 at

PSORS5 confirms a role in susceptibility to Psoriasis vulgaris. The Journal of Investigative Dermatology

125: 906-912.

Hughes J.M., Murad F., Chang B., Guerrant R.L. (1978): Role of cyclic GMP in the action of heat-stable

enterotoxin of Escherichia coli. Nature 271: 755-756.

Page 94: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 93 -

Huijsdens X.W., Linskens R.K., Mak M., Meuwissen S.G.M., Van Den Broucke-Grauls C.M.J.E.,

Savelkoul P.H.M. (2002): Quantification of Bacteria Adherent to Gastrointestinal Mucosa by Real-Time

PCR. Clinical Microbiology 40: 4423-4427.

Humphray S.J., Scott C.E., Clark R., Marron B., Bender C., Camm N., Davis J., Jenks A., Noon A., Patel

M., Sehra H., Yang F., Rogatcheva M.B., Milan D., Chardon P., Rohrer G., Nonneman D., De Jong P.,

Meyers S.N., Archibald A., Beever J.E., Schook L.B., Rogers J. (2007): A high utility integrated map of

the pig genome. Genome Biology 8: R139.

Jacobsen M. (2011): Genetic dissection of the ETEC F4ab/ac susceptibility locus in pigs. PhD thesis,

ISBN 978-87-7611-423-7.

Jacobsen M., Kracht S.S., Esteso G., Cirera S., Edfors I., Archibald A.L., Bendixen C., Andersson L.,

Fredholm M., Jørgensen C.B. (2010): Refined candidate region specified by haplotype sharing for

Escherichia coli F4ab/F4ac susceptibility alleles in pigs. Animal Genetics 41: 21-25.

Jensen B.B. (1998): The impact of feed additives on the microbial ecology of the gut in young pigs.

Journal of Animal and Feed Sciences 7: 45-64.

Ji H., Ren J., Yan X., Huang X., Zhang B., Zhang Z., Huang L. (2011): The porcine MUC20 gene:

molecular characterization and its association with susceptibility to enterotoxigenic Escherichia coli

F4ab/ac. Molecular Biology Reports 38: 1593-1601.

Joller D. (2009): Comparative molecular approaches to identify host determinants mediating adhesion of

E. coli F4 strains in pigs. Dissertation ETH Zurich number 18518.

Joller D., Jørgensen C.B., Bertschinger H.U., Python P., Edfors I., Cirera S., Archibald A.L., Bürgi E..,

Karlskov-Mortesen P., Andersson L., Fredholm M., Vögeli P. (2009): Refined localization of the

Escherichia coli F4ab/F4ac receptor locus on pig chromosome 13. Animal Genetics 40: 749-752.

Jones G.W. & Rutter J.M. (1972): Role of the K88 antigen in the pathogenesis of neonatal diarrhea

caused by Escherichia coli in piglets. Infection and Immunity 6: 918-927.

Jørgensen C.B., Cirera S., Andersson S.I., Archibald A.L., Raudsepp T., Chowdhary B., Edfors-Lilja I.,

Andersson L., Fredholm M. (2003): Linkage and comparative mapping of the locus controlling

susceptibility towards E. coli F4ab/ac diarrhoea in pigs. Cytogenetic and Genome Research 102: 157-162.

Jørgensen C.B., Cirera S., Archibald A.L., Andersson S.I., Fredholm M., Edfors-Lilja I. (2004): Porcine

polymorphisms and methods for detecting them. Patent number WO2004048606.

Page 95: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 94 -

Kapitany R.A., Scoot A, Forsyth G.W., McKenzie S.L., Worthington R.W. (1979): Evidence for Two

Heat-Stable Enterotoxins Produced by Enterotoxigenic Escherichia coli. Infection and Immunity 24: 965-

966.

Karlskov-Mortensen P., Hu Z.L., Gorodkin J., Reecy J.M., Fredholm M. (2007): Identification of 10882

porcine microsatellite sequences and virtual mapping of 4528 of these sequences. Animal Genetics 38:

401-405.

Kim Y.J., Kim J.H., Hur J., Lee J.H. (2010): Isolation of Escherichia coli from piglets in South Korea

with diarrhea and characteristics of the virulence genes. Canadian Journal of Veterinary Research 74: 59-

64.

Kjaersgard H.D., Christensen G., Svensmark B., Bækbo P. (2002): Mortality in pigs. Proceedings of the

17th IPVS Congress. Ames, Iowa, USA.

Lang T., Hansson G.C., Samuelsson T. (2006): An inventory of mucin genes in the chicken genome

shows that the mucin domain of MUC13 is encoded by multiple exons and that ovomucin is part of a

locus of related gel-forming mucins. BMC Genomics 7: 197.

Lee R.W., Strommer J., Hodgins D., Shewen P.E., Niu Y., Lo R.Y. (2001): Towards development of an

edible vaccine against bovine pneumonic pasteurellosis using transgenic white clover expressing a

Mannheimia Haemolytica A1 leukotoxin 50 fusion protein. Infection and Immunity 69: 5786-5793.

Li Y., Qiu X. & Li H., Zhang Q. (2007): Adhesive patterns of Escherichia coli F4 in piglets of three

breeds. Journal of Genetics and Genomics 34: 591-599.

Li X.Y., Jin L.J., Uzonna J.E., Li S.Y., Liu J.J., Li H.Q., Lu Y.N., Zhen Y.H., Xu Y.P. (2009): Chitosan-

alginate microcapsules for oral delivery of egg yolk immunoglobulin (IgY): In vivo evaluation in a pig

model of enteric colibacillosis. Veterinary Immunology and Immunopathology 129: 132-136.

Liang W., Huang Y., Yang X., Zhou Z., Pan A., Qian B., Huang C., Chen J., Zhang D. (2006): Oral

immunization of mice with plant-derived fimbrial adhesion FaeG induces systemic and mucosal K88ad

enterotoxigenic Escherichia coli-specific immune responses. FEMS Immunology & Medical

Microbiology 46: 393-399.

Luther H., Vögeli P., Hofer A. (2009): Increasing genetic E. coli F18 resistance in Swiss pigs.

http://www.eaap.org/Barcellona/Book_Abstracts.pdf.

Mably J.D., Mohideen M.A., Burns C.G., Chen J.N., Fishman M.C. (2003): Heart of glass regulates the

concentric growth of the heart in zebrafish. Current Biology 13: 2138-2147.

Page 96: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 95 -

MacKinnon J.D. (1998): Enteritis in the young pig caused by Escherichia coli. The Pig Journal 41: 227-

255.

Madoroba E., Van Driessche E., De Greve H., Mast J., Ncube I., Read J., Beeckmans S. (2009):

Prevalence of enterotoxigenic Escherichia coli virulence genes from scouring piglets in Zimbabwe.

Tropical Animal Health and Production 41: 1539-1547.

Maher D.M., Gupta B.K., Nagata S., Jaggi M., Chauhan S.C. (2011): Mucin 13: structure, function, and

potential roles in cancer pathogenesis. Molecular Cancer Research 9: 531-537.

Marin, F., Luquet, G., Marie, B., Medakovic, D. (2008): Molluscan Shell Proteins: Primary Structure,

Origin, and Evolution. Current Topics in Developmental Biology 80: 209-276.

Markwalder K. (2001): Travelers' diarrhea. Therapeutische Umschau 58: 367-371.

Mathie R.T. (2003): Clinical outcomes research: contributions to the evidence base for homeopathy.

Homeopathy 92: 56-57.

McCluskie M.J. & Davis H.L. (2000): Mucosal immunization with DNA vaccines. Methods in Molecular

Medicine 29: 287-295.

Meijerink E., Fries R., Vögeli P., Masabanda J., Wigger G., Stricker C., Neuenschwander S.,

Bertschinger H.U., Stranzinger G. (1997): Two alpha(1, 2) fucosyltransferase genes on porcine

chromosome 6q11 are closely linked to the blood group inhibitor (S) and Escherichia coli F18 receptor

(ECF18R) loci. Mammalian Genome 8: 736-741.

Meijerink E., Neuenschwander S., Fries R., Dinter A., Bertschinger H.U., Stranzinger G., Vögeli P.

(2000): A DNA polymorphism influencing alpha(1, 2) fucosyltransferase activity on the pig FUT1

enzyme determines susceptibility of small intestinal epithelium to Escherichia coli F18 adhesion.

Immunogenetics 52: 129-136.

Michaels R.D., Whipp S.C., Rothschild M.F. (1994): Resistance of Chinese Meishan, Fengjing, and

Minzhu pigs to the K88ac+ strain of Escherichia coli. American Journal of Veterinary Research 55: 333-

338.

Moon H.W. (1978): Mechanisms in the pathogenesis of diarrhea: a review. Journal of the American

Veterinary Medical Association 172: 443-448.

Moon H.W. (1997): Comparative histopathology of intestinal infections. Advances in Experimental

Medicine and Biology 412: 1-19.

Page 97: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 96 -

Moore, W.E. & Holdeman L.V. (1974): Special problems associated with the isolation and identification

of intestinal bacteria in fecal flora studies. The American Journal of Clinical Nutrition 27: 1450-1455.

Mueller O., Hahnenberger K., Dittmann M., Yee H., Dubrow R., Nagle R., Ilsley D. (2000): A

microfluidic system for high-speed reproducible DNA sizing and quantitation. Electrophoresis 21: 128-

134.

Nagy B., Casey T.A., Moon H.W. (1990): Phenotype and genotype of Escherichia coli isolated from pigs

with postweaning diarrhea in Hungary. Journal of Clinical Microbiology 28: 651-653.

Nagy B. & Fekete P.Z. (2005): Enterotoxigenic Escherichia coli in veterinary medicine. International

Journal of Medical Microbiology 295: 443-454.

Nataro J.P. & Kaper J.B. (1998): Diarrheagenic Escherichia coli. Clinical Microbiology Reviews 11:

142-201.

Ngeleka M., Pritchard J., Appleyard G., Middleton D.M. Fairbrother J.M. (2003): Isolation and

association of Escherichia coli AIDA-I/STb, rather than EAST1 pathotype, with diarrhea in piglets and

antibiotic sensitivity of isolates. Journal of Veterinary Diagnostic Investigation 15: 242-252.

NOAH, National Office of Animal Health (2001): Antibiotics for animals.

http://www.noah.co.uk/issues/antibiotics.htm.

Owczarzy R., Tataurov A.V., Wu Y., Manthey J.A., McQuisten K.A., Almabrazi H.G., Pedersen K.F.,

Lin Y., Garretson J., McEntaggart N.O., Sailor C.A., Dawson R.B., Peek A.S. (2008): IDT SciTools: a

suite for analysis and design of nucleic acid oligomers. Nucleic Acids Research 36: W163-W169.

Ojeniyi B., Ahrens P., Meyling A. (1994): Detection of fimbrial and toxin genes in Escherichia coli and

their prevalence in piglets with diarrhea. The Application of colony hybridization assay, polymerase chain

reaction and phenotypic assays. Journal of Veterinary Medicine Series B 41: 49-59.

Ouyang J., Zeng W., Ren J., Yan X., Zhang Z., Yang M., Han P., Huang X., Ai H., Huang L. (2012):

Association of B3GNT5 polymorphisms with susceptibility to ETEC F4ab/ac in the white Duroc ×

Erhualian intercross and 15 outbred pig breeds. Biochemical Genetics 50: 19-33.

Panaro N.J., Yuen P.K., Sakazume T., Fortina P., Kricka L.J., Wilding P. (2000): Evaluation of DNA

fragment sizing and quantification by the Agilent 2100 Bioanalyzer. Clinical Chemistry 46: 1851-1853.

Peng Q.L., Ren J., Yan X.M., Huang X., Tang H., Wang Y.Z., Zhang B., Huang L.S. (2007): The

g.243A>G mutation in intron 17 of MUC4 is significantly associated with susceptibility/resistance to

ETEC F4ab/ac infection in pigs. Animal Genetics 38: 397-400.

Page 98: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 97 -

Pentsuk N. & Van Der Laan J.W. (2009): An interspecies comparison of placental antibody transfer: new

insights into developmental toxicity testing of monoclonal antibodies. Birth Defects Research. Part B:

Developmental and Reproductive Toxicology 86: 328-344.

Python P., Jörg H., Neuenschwander S., Hagger C., Stricker C., Bürgi E., Bertschinger H.U., Stranzinger

G., Vögeli P. (2002): Fine-mapping of the intestinal receptor locus for enterotoxigenic Escherichia coli

F4ac on porcine chromosome 13. Animal Genetics 33: 441-447.

Python P. (2003): Genetic host determinants associated with the adhesion of E. coli with fimbriae F4 in

swine. Dissertation, ETH Zurich.

Python P., Jorg H., Neuenschwander S., Asai-Coakwell M., Hagger C., Burgi E., Bertschinger H.U.,

Stranzinger G., Vögeli P. (2005): Inheritance of the F4ab, F4ac and F4ad E. coli receptors in swine and

examination of four candidate genes for F4acR. Journal of Animal Breeding and Genetics 122: 5-14.

Qadri F., Svennerholm A.M., Faruque A.S.G., Sack R.B. (2005): Enterotoxienic Escherichia coli in

developing countries: epidemiology, microbiology, clinical features, treatment, and prevention. Clinical

Microbiology Reviews 18: 465-483.

Ramos A.M., Crooijmans R.P., Affara N.A., Amaral A.J., Archibald A.L., Beever J.E., Bendixen C.,

Churcher C., Clark R., Dehais P., Hansen M.S., Hedegaard J., Hu Z.L., Kerstens H.H., Law A.S., Megens

H.J., Milan D., Nonneman D.J., Rohrer G.A., Rothschild M.F., Smith T.P., Schnabel R.D., Van Tassell

C.P., Taylor J.F., Wiedmann R.T., Schook L.B., Groenen M.A. (2009): Design of a high density SNP

genotyping assay in the pig using SNPs identified and characterized by next generation sequencing

technology. PloS One 4: e6524.

Rampoldi A., Jacobsen M.J., Bertschinger H.U., Joller D., Bürgi E., Vögeli P., Andersson L., Archibald

A.L., Fredholm M., Jørgensen C.B., Neuenschwander S. (2011): The receptor locus for Escherichia coli

F4ab/F4ac in the pig maps distal to the MUC4-LMLN region. Mammalian Genome 22: 122-129.

Rapacz J. & Hasler-Rapacz J. (1986): Polymorphism and inheritance of swine small intestinal receptors

mediating adhesion of three serological variants of Escherichia coli producing K88 pilus antigen. Animal

Genetics 17: 305-321.

Rasschaert K., Verdonck F., Goddeeris B.M., Duchateau L., Cox E. (2007): Screening of pigs resistant to

F4 enterotoxigenic Escherichia coli (ETEC) infection. Veterinary Microbiology 123: 249-253.

Reid G., Howard J., Gan B.S. (2001): Can bacterial interference prevent infection? Trends in

Microbiology 9: 424–428.

Page 99: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 98 -

Ren J., Yan X., Ai H., Zhang Z., Huang X., Ouyang J., Yang M., Yang H., Han P., Zeng W., Chen Y.,

Guo Y., Xiao S., Ding N., Huang L. (2012): Susceptibility towards enterotoxigenic Escherichia coli F4ac

diarrhea is governed by the MUC13 gene in pigs. PLoS One 7: e44573.

Rohrer G.A., Alexander L.J., Keele J.W., Smith T.P., Beattie C.W. (1994): A Microsatellite Linkage Map

of the porcine Genome. Genetics 136: 231-245.

Ronaghi M., Uhlén M., Nyrén P. (1998): A sequencing method based on real-time pyrophosphate.

Science 281: 363-365.

Roselli M., Finamore A., Britti M.S., Konstantinov S.R., Smidt H., De Vos W.M., Mengheri E. (2007):

The novel porcine Lactobacillus Sobrius strain protects intestinal cells from enterotoxigenic Escherichia

coli K88 infection and prevents membrane barrier damage. Journal of Nutrition 137: 2709-2716.

Rosengren L.B., Waldner C.L., Reid-Smith R.J., Checkley S.L., McFall M.E., Rajić A. (2008):

Antimicrobial resistance of fecal Escherichia coli isolated from grow-finish pigs in 20 herds in Alberta

and Saskatchewan, Canada. Canadian Journal of Veterinary Research 72: 160-167.

Rozen S. & Skaletsky H.J. (2000): Primer3 on the WWW for general users and for biologist

programmers. In Krawets S. & Misener S. (editors): Bioinformatics-methods and protocols: methods in

molecular biology. Humana Press, Totowa, 1st edition: 365-386.

Sarrazin E., Fritzsche C., Bertschinger H.U. (2000): Hauptvirulenzfaktoren bei Escherichia coli-Isolaten

von über zwei Wochen alten Schweinen mit Ödemkrankheit und/oder Colidiarrhöe. Schweizer Archiv

Tierheilk 142: 625-630.

Schook L.B., Beever J.E., Rogers J., Humphray S., Archibald A., Chardon P., Milan D., Rohrer G.,

Eversole K. (2005): Swine Genome Sequencing Consortium (SGSC): a strategic roadmap for sequencing

the pig genome. Comparative and Functional Genomics 6: 251-255.

Schroyen M., Stinckens A., Verhelst R., Geens M., Cox E., Niewold T., Buys N. (2012): Susceptibility of

piglets to enterotoxigenic Escherichia coli is not related to the expression of MUC13 and MUC20.

Animal Genetics 43: 324-327.

Sears C.L. (2005): A dynamic partnership: celebrating our gut flora. Anaerobe 11: 247-255.

Sellwood R., Gibbons R.A., Jones G.W., Rutter J.M. (1975): Adhesion of enteropathogenic Escherichia

coli to pig intestinal brush borders: the existence of 2 pig phenotypes. Journal of Medical Microbiology 8:

405-411.

Page 100: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 99 -

Sellwood R. (1979): Escherichia coli diarrhea in pigs with or without the K88 receptor. Veterinary

Record 105: 228-230.

Sellwood R. (1982): Escherichia coli associated porcine neonatal diarrhea: Antibacterial activities of

colostrum from genetically susceptible and resistant sows. Infection and Immunity 35: 396-401.

Shahriar F., Ngeleka M., Gordon J.R., Simko E. (2006): Identification by mass spectroscopy of F4ac-

fimbrial-binding proteins in porcine milk and characterization of lactadherin as an inhibitor of F4ac-

positive Escherichia coli attachment to intestinal villi in vitro. Developmental and Comparative

Immunology 30: 723-734.

Simon G.L. & Gorbach S.L. (1986): The human intestinal microflora. Digestive Diseases and Sciences

31: 147S-162S.

Smith M.G., Jordan D., Chapman T.A., Chin J.J.-C., Barton M.D., Do T.N., Fahy V.A., Fairbrother J.M.,

Trott D.J. (2010): Antimicrobial resistance and virulence gene profiles in multi-drug resistant

enterotoxigenic Escherichia coli isolated from pigs with post-weaning diarrhoea. Veterinary

Microbiology 145: 299-307.

Snodgrass D.R., Chandler D.S., Makin T.J. (1981): Inheritance of Escherichia coli K88 adhesion in pigs:

identification of nonadhesion phenotypes in a commercial herd. The Veterinary Record 109: 461-463.

Steinhoff U. (2005): Who controls the crowd? New findings and old questions about the intestinal

microflora. Immunology Letters 99: 12-16.

Steemers F.J. & Gunderson K.L. (2007): Whole genome genotyping technologies on the BeadArray

platform. Biotechnology Journal 2: 41-49.

Sterzl J., Rejnek J., Tràvnìcek J. (1966): Impermeability of pig placenta for antibodies. Folia

Microbiologica 11: 7-10.

Sweeney E.J. (1968): Escherichia coli enteric disease of swine: observation on herd resistance. Irish

Veterinary Journal 22: 42-46.

Thiele H. & Nürnberg P. (2005): HaploPainter: a tool for drawing pedigrees with complex haplotypes.

Bioinformatics 21:1730-1732.

Thorns C.J., Boarer C.D.H., Morris J.A. (1987): Production and evaluation of monoclonal-antibodies

directed against the K88 fimbrial adhesin produced by Escherichia coli enterotoxigenic for piglets.

Research in Veterinary Science 43: 233-238.

Page 101: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 100 -

Toledo A., Gómez D., Cruz C., Carreón R., López J., Giono S., Castro A.M. (2012): Prevalence of

virulence genes in Escherichia coli strains isolated from piglets in the suckling and weaning period in

Mexico. Journal of Medical Microbiology 61: 148-156.

Travers K.J., Chin C.S., Rank D.R., Eid J.S., Turner S.W. (2010): A flexible and efficient template format

for circular consensus sequencing and SNP detection. Nucleid Acids Research 38: e159.

Turner M. (2011): German E. coli outbreak caused by previously unknown strain. Nature

doi:10.1038/news.2011.345.

Valpotic I., Frankovic M., Vrbanac I. (1992): Identification of infant and adult swine susceptible to

enterotoxigenic Escherichia coli by detection of receptors for F4(K88)ac fimbriae in brush borders or

feces. Comparative Immunology, Microbiology and Infectious Diseases 15: 271-279.

Vandamme K., Melkebeek V., Cox E., Remon J.P., Vervaet C. (2011): Adjuvant effect of Gantrez®AN

nanoparticles during oral vaccination of piglets against F4+ enterotoxigenic Escherichia coli. Veterinary

Immunology and Immunopathology 139: 148-155.

Van Den Broeck W., Cox E., Goddeeris B.M. (1999): Receptor-dependent immune responses in pigs

after oral immunization with F4 fimbriae. Infection and Immunity 67: 520-526.

Van Den Broeck W., Cox E., Oudega B., Goddeeris B.M. (2000): The F4 fimbrial antigen of Escherichia

coli and its receptors. Veterinary Microbiology 71: 223-244.

Vögeli P., Bolt R., Fries R., Stranzinger G. (1994): Co-segregation of the malignant hyperthermia and the

Arg(615)-Cys(615) mutation in the skeletal-muscle calcium-release channel protein in 5 European

Landrace and Pietrain pig breeds. Animal Genetics 25: 59-66.

Vögeli P., Bertschinger H.U., Stamm M., Stricker C., Hagger C., Fries R., Rapacz J., Stranzinger G.

(1996): Genes specifying receptors for F18 fimbriated Escherichia coli, causing oedema disease and

postweaning diarrhoea in pigs, map to chromosome 6. Animal Genetics 27: 321-328.

Vogt R.L. & Dippold L. (2005): Escherichia coli O157:H7 outbreak associated with consumption of

ground beef, June–July 2002. Public Health Reports 120: 174–178.

Wagner B., Hufnagl K., Radauer C., Wagner S., Baier K., Scheiner O., Wiedermann U., Breiteneder H.

(2004): Expression of the B subunit of the heat-labile enterotoxin of Escherichia coli in tobacco mosaic

virus-infected Nicotiana benthamiana plants and its characterization as mucosal immunogen and

adjuvant. Journal of Immunological Methods 287: 203-215.

Page 102: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 101 -

Wang Y., Ren J., Lan L., Yan X., Huang X., Peng Q., Tang H., Zhang B., Ji H., Huang L. (2007):

Characterization of polymorphisms of transferrin receptor and their association with susceptibility to

ETEC F4ab/ac in pigs. Journal of Animal Breeding and Genetics 124: 225-229.

Westerman R.B., Mills K.W., Phillips R.M., Fortner G.W., Greenwood J.M. (1988): Predominance of the

ac variant in K88-positive Escherichia coli isolates from swine. Journal of Clinical Microbiology 26: 149-

150.

Williams S.J., Wreschner D.H., Tran M., Eyre H.J., Sutherland G.R., McGuckin M.A. (2001): MUC13, a

novel human cell surface mucin expressed by epithelial and hemopoietic cells. The Journal of Biological

Chemistry 276: 18327-18336.

Winterø A.K. & Fredholm M. (1995): Three porcine polymorphic microsatellite loci (S0075, S0151,

S0158). Animal Genetics 26: 125-126.

Wittig W., Klie H., Gallien P., Lehmann S., Timm M., Tschäpe H. (1995): Prevalence of the fimbrial

antigens F18 and K88 and of enterotoxins and verotoxins among Escherichia coli isolated from weaned

pigs. International Journal of Medical Microbiology 283: 95-104.

Yan X., Huang X., Ren J., Zou Z., Yang S., Ouyang J., Zeng W., Yang B., Xiao S., Huang L. (2009):

Distribution of Escherichia coli F4 adhesion phenotypes in pigs of 15 Chinese and Western breeds and a

White Duroc x Erhualian intercross. Journal of Medical Microbiology 58: 1112-1117.

Zhang W., Berberov E.M., Freeling J., He D., Moxley R.A., Francis D.H. (2006): Significance of heat-

stable and heat-labile enterotoxins in porcine colibacillosis in an additive model for the pathogenicity

studies. Infection and Immunity 74: 3107-3114.

Zhang W., Zhao M., Ruesch L., Omot A., Francis D. (2007): Prevalence of virulence genes in

Escherichia coli strains recently isolated from young pigs with diarrhea in the U.S. Veterinary

Microbiology 123: 145-152.

Zhang B., Ren J., Yan X., Huang X., Ji H., Peng Q., Zhang Z., Huang L. (2008): Investigation of the

porcine MUC13 gene: isolation, expression, polymorphisms and strong association with susceptibility to

enterotoxigenic Escherichia coli F4ab/ac. Animal Genetics 39: 258-266.

Zhao L., Chen X., Xu X., Song G., Liu X. (2009): Analysis of the AIDA-I gene sequence and prevalence

in Escherichia coli isolates from pigs with post-weaning diarrhoea and oedema disease. Veterinary

Journal 180: 124-129.

Page 103: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 102 -

ABBREVIATIONS

Description

A,T,G,C Adenine, thymine, guanine, cytosine

ADP Adenosine diphosphate

ATP Adenosine triphosphate

BAC Bacterial artificial chromosome

bp Base pair

°C Degree Celsius

cDNA Complementary deoxyribonucleic acid

cM CentiMorgan

ddH2O Double distilled H2O

DMSO Dimethyl sulfoxide

DNA Deoxyribonucleic acid

dNTP Deoxynucleotide triphosphate

EDTA Ethylenediaminetetraacetic acid

E. coli Escherichia coli

ETEC Enterotoxigenic Escherichia coli

EtOH Ethanol

F4 Fimbrial antigen F4, former K88

F4ab, F4ac, F4ad Fimbriae of type F4 with ab, ac and ad antigens

F18 Fimbrial antigen F18

FaeG Major subunit protein of F4 fimbriae

g Gram

x g Acceleration of gravity

h Hour

H2O Water

HSA Homo Sapiens chromosome

Kb Kilobase (103 bp)

kDa KiloDalton (103 Dalton)

l Liter

LT-I, LT-II Heat-labile enterotoxin type I and type II

M Molarity (mol/liter)

Mb Megabase (106 bp)

mg Milligram (10-3 gram)

µg Microgram (10-6 gram)

min Minute

ml Milliliter (10 -3 liter)

µl Microliter (10-6 liter)

mmol Millimol (10-3 mol)

mol Mole 6 x 1023 molecules

Page 104: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

- 103 -

Continued from previous page

Description

mRNA Messenger RNA

NaAc Natrium acetate

NaCl Sodium chloride

NaOH Sodium hydroxide

NEH Nordic Experimental Herd

ng Nanogram (10-9 gram)

nl Nanoliter (10-9 liter)

PBS Phosphate buffered saline

PCR Polymerase chain reaction

RFLP Restriction fragment length polymorphism

RNA Ribonucleic acid

RNase(s) Ribonuclease(s)

RNasin RNases inhibitor

rpm Revolutions per minute

rRNA Ribosomal RNA

RT-PCR Reverse transcription PCR

s Second

SDS Sodium dodecyl sulfate

SNP Single nucleotide polymorphism

SPS Swiss Performing Station

SSC Saline sodium citrate buffer

SSC Sus Scrofa chromosome

STa, STb Heat-stabile enterotoxin a and b

Ta Annealing temperature

TBE Tris-borate-EDTA buffer

U Unit

UEH University experimental herd

5’ UTR Region at 5’ of a transcript, before the start codon, untranslated

3’ UTR Region at 3’ of a transcript, after the stop codon, untranslated

v/v Volume per volume

w/v Weight per volume

List of figures

Figure 1.1 E. coli bacterium with fimbriae. 13

Figure 1.2 Small intestinal brush border with strong ETEC F4 adhesion. 14

Figure 1.3 Gene cluster encoding F4 fimbriae. 16

Figure 1.4 Location of candidate genes for F4bcR on SSC13. 24

Page 105: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

SUMMARY

104

Continued from previous page

Figure 2.1 Determination of the ETEC F4 receptor phenotype in the MAT 31

Figure 2.2 Example of a pyrogram 42

Figure 2.3 Simplified diagram of the Illumina HiSeq 2000 sequencing procedure 45

Figure 2.4 Simplified diagram of the PacBio RS sequencing procedure 46

Figure 2.5 Electropherograms of total RNA extracted from intestinal scrapings 49

Figure 2.6 Simplified diagram of the SOLiDTM 3 System RNA sequencing procedure 51

Figure 3.1:

Left: Restriction pattern of DdeI digestion of MUC4-6138-F/6887-R 749 bp PCR product to

determine the MUC4_g.6242 A>G polymorphism

Middle: Restriction pattern of Hin1II digestion of MUC4-7741-F/8202-R PCR products to

determine the MUC4_g.7947 G>A polymorphism

Right: Restriction pattern of XbaI digestion of MUC4-8012-F/8378-R 367 bp PCR product

to determine the MUC4_g.8227 G>C polymorphism

54

Figure 3.2 Diagram of three-generation family tree of 2349 and haplotypes in the SSC13q41-q44 region 55

Figure 3.3:

Left: Restriction pattern of TaqI digestion of TNK2e6-7-Fc2/R PCR product to determine the

TNK2_g.7075 A>C polymorphism

Middle: Restriction pattern of BseDI digestion of TNK2e9j-F/R PCR product to determine

the TNK2_g.7717 T>C polymorphism

Right: Restriction pattern of AluI digestion of TNK2e12b-Fv2/Rv2 PCR product to

determine the TNK2_g.11142 A>G polymorphism

58

Figure 3.4

Left: Restriction pattern of TspRI digestion of HEG1-F/R PCR product to determine the

HEG1_g.5244 T>C polymorphism

Middle: Restriction pattern of FOKI digestion of MUC13-F/R PCR product to determine the

MUC13-226 A>G polymorphism

Right: Restriction pattern of HaeIII digestion of MUC13-ED2-F/R PCR product to determine

the MUC13_c1930 T>G polymorphism

59

Figure 3.5 Diagram of a second three-generation family tree of 2349 and haplotypes in the SSC13q41-

q44 region 62

Figure 3.6 Family tree and haplotypes of sow 9480XJZ in the MUC4-MARC0045417 region 66

Figure 3.7 Alternative splicing of gene HEG1 in exon 5 69

Figure 3.8 Position of long-range PCR products in the HEG1-ITGB5 interval 71

Figure 3.9 SNPs positions in the HEG1-ITGB5 interval 72

List of tables

Table 1.1 Phenotypes observed in pigs according to the binding of ETEC F4 variants 18

Table 2.1 Primers for standard PCR 34

Table 2.2.1 Primers for long-range PCR in the intergenic region HEG1-MUC13 38

Table 2.2.2 Primers for long-range PCR in the intergenic region MUC13-ITGB5 39

Table 2.3 Primers for pyrosequencing 40

Table 2.4 Microsatellite markers used for genescan analyses 41

Page 106: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

SUMMARY

105

Continued from previous page

Table 2.5 Selected SNPs belonging to the Illumina Porcine SNP60 BeadChip 43

Table 2.6 Selected SNPs in TNK2, MUC4, HEG1, and MUC13 genes and in the Illumina Porcine

SNP60 BeadChip 44

Table 3.1 Polymorphisms in genes ZDHHC19, TNK2, MUC4, KIAA0226, LRCH3, LMLN and MUC13

of 2349’s family 57

Table 3.2 List of SNPs from the Porcine SNP60 DNA BeadChip located in the interval ZDHHC19-

S0075 60

Table 3.3 Haplotypes of the six pigs recombinant in MUC4-F4bcR from the SPS 64

Table 3.4 Genotyping of the 46 pigs selected from the SPS in markers MUC4_g.8227, ALGA0072075,

KVL1293, SNP 1, SNP 2, ALGA0106330, MUC13-226 and MUC13-813 74

Table 3.5 Phenotypes observed in the 489 pigs from UEH according to the binding of ETEC F4

variants 75

Table 3.6 E. coli F4ad adhesion strengths at four intestinal sites in pigs of phenotypes E1, E2 and R 76

Table 3.7 Distribution of the three phenotypes for ETEC F4d in progeny with known phenotyped

parents 77

Table 3.8 Influence of phenotypes of parents on the median adhesion strengths (± SD) at the four

intestinal sites A through D of the progeny 78

Table 3.9 Kruskal-Wallis test with the six mating combinations of the three phenotypes for ETEC F4ad 78

Table 3.10 Evaluation of the Pedigree Analysis Package models 80

Table 3.11 Analyses of the largest litters (≥12 pigs) from UEH 81

Appendix

Media and solutions

Agarose gel 0.8-1.5% (w/v) PBS buffer

TBE 1 X NaCl 145 Mm

Agarose 0.8-1.5% Na2HPO4 9 Mm

Ethidium bromide 0.1 µg/ml NaH2PO4 1.3 mM

DMSO-Hanks-Medium (as in Bosi et al., 2004) PBS-EDTA buffer

Hanks’ Balanced Salt Solution 80 ml NaCl 96 Mm

Fetal Calf Serum 10 ml Na2HPO4 5.5 mM

DMSO 10 ml KH2PO4 1.5 mM

Glycerol 30 ml KCl 1.5 mM

BSA 1 g EDTA 10 mM

DNA loading dye-Bromophenol blue PBS-formaldehyde

Bromophenol blue 0.25% (w/v) Formaldehyde solution 2% (v/v)

D(+)-sucrose 40% (w/v) PBS buffer 1X 98% (v/v)

Page 107: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

SUMMARY

106

Continued from previous page

DNA loading dye-XCFF Polyacrylamide gel 4.5%

XCFF 0.25% (w/v) ddH2O 23 ml

Orange G 0.26% (w/v) Urea 18 g

D(+)-sucrose 40% (w/v) TBE buffer 10 X 5 ml

Acrylamide: bis-acryl-amide (29:1) solution 7.5 ml

Formamide loading dye TEMED 15 µl

Formamide 4 X Ammonium persulfate 10% solution 350 µl

Loading buffer 1 X

Silica suspension 100 mg/ml

Lysis Buffer Silicon-dioxide 10 g

Sucrose 320 mM PBS buffer 1 X 100 ml

Tris-HCl 10mM

MgCl2 5mM TBE buffer 10X

Triton X-100 1% (w/v) Tris-HCL 890 mM

H3BO3 890 mM

Mannose buffer 2% EDTA 20 mM

D(+)-Mannose 2% (w/v)

PBS buffer 1X 98% (w/v) TE buffer

Tris-HCl 10 mM

ECL Buffer EDTA 1 mM

NH4Cl 155 mM

KHCO3 10 mM Wash-buffer

EDTA 0.1 mM NaCl 50 mM

Tris-HCl 10 mM

EDTA 2.5 mM

EtOH 50% (v/v)

Chemicals

Product Producer

λ-phage DNA 500 µg/ml GE

100 bp ladder GE

100 bp ladder direct load Sigma

50 bp ladder GE

50 bp ladder direct load Sigma

1 Kb ladder Life Technologies

Acrylamide: bis-acryl-amide (29:1) solution Bio-Rad

Agarose low EEO Sigma

BigDye sequencing mix AB

Columbia sheep blood agar Oxoid

DNase, RNase-free Qiagen

dNTPs, 100 mM GE

Page 108: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

SUMMARY

107

Continued from previous page

Product Producer

dNTPs, 100 mM Sigma

Ethanol (EtOH) Merck

Etidium bromide (EtBr)

Formamide Fluka

Formaldehyde solution Sigma

Genescan 350 TAMRA or ROX size standard AB

Guanidine HCl Sigma

Isopropanol Merck

M-MLV reverse transcription system Promega

SequalPrep Long PCR kit with dNTPs Invitrogen

Silicon-dioxide Sigma

Sodium Acetate Fluka

Taq JumpStart DNA polymerase 2.5 U/µl Sigma

TRIzol Invitrogen

Trypticase soy broth (TSB) Becton Dickinson

Restriction enzymes

Producer

AluI Fermentas

BseDI (SecI) Fermentas

DdeI BioLabs

DdeI (HpyF3I) Fermentas

DdeI Promega

FOKI BioLabs

HaeIII Fermentas

Hin1II (NlaIII) Fermentas

PstI Boehringer Mannheim

TaqI Roche

TspRI BioLabs

TspRI (TscAI) Fermentas

XbaI Fermentas

XbaI Promega

Labware

Product Producer

6-well macroplates 82 x 127 mm Greiner

6-well macroplates 82 x 127 mm Orange

Blood tubes 10 ml with EDTA Vacuette Greiner

Page 109: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

SUMMARY

108

Continued from previous page

Product Producer

Blood tubes 10 ml with EDTA Venosafe Terumo

Centrifuge tubes 15 ml Falcon

Centrifuge tubes 50 ml TPP

Cover glass 18 x 18 mm Menzel

Cryotubes 1.8 ml Nunc

Genelute Mammalian Genomic DNA miniprep kit Sigma

Glass slide frosted ends 76 x 26 mm Menzel

Micro tubes 1.5 ml Treff

Micro tubes 2 ml Treff

Microcon centrifugal filter units Millipore

Montage PCR centrifugal filter devices Millipore

PCR 8-strip tubes SSI

PCR 8-strip tubes VWR

PCR plates 96 well Axygen

PCR plates 96 well ABI

PCR plates 96 well low profile Thermo

PCR tubes 0.2 ml SSI

PCR tubes 0.5 ml Axygen

Petri dishes 92 mm Falcon

QIAquick Gel extraction kit Qiagen

Serological pipette 1 ml Falcon

Serological pipette 5 ml Falcon

Serological pipette 5 ml Sarstedt

Serological pipette 10 ml Falcon

Serological pipette 25 ml VWR

Tips 10 µl Axygen

Tips 30 µl Matrix

Tips 200 µl Treff

Tips 1000 µl Treff

Wide-necked bottles PVC 100 ml Semadeni

Page 110: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

SUMMARY

109

SNPs Nomenclature

All the SNPs discovered in this project have been deposited in dbSNP (NCBI) under batch

number CHCF. The SNPs are given with their dbSNP ID and position according to the reference

sequence Sscrofa 10.2.

SNPs NCBI

dbSNP ID

Position on

Sscrofa 10.2

SNP 1 CHCF1 144944384

SNP 2 CHCF3 144978031

LMLN_g.15920 CHCF5 144201963/1442551221

ZNF148_g.96828 CHCF6 144544700

SLC12A8_g.1365 CHCF7 144499531

SLC12A8_g.22157 CHCF8 144631142

SLC12A8_g.129502 CHCF9 144853219

SLC12A8_c.1990 CHCF10 144853282

SLC12A8_c.2947 CHCF11 144849048

HEG1_g.5244 CHCF12 144823489

HEG1_c.321 CHCF13 144800514

HEG1_c.1905 CHCF14 144783551

HEG1_c.1917 CHCF15 144783539

HEG1_g.49379 CHCF16 144779899

HEG1_c.6955 CHCF17 144707761/1449312421

JN613413_23529 CHCF18 144935458

JN613413_34623 CHCF19 144946537

JN613413_34996 CHCF20 144946910

JN613413_35253 CHCF21 144947167

JN613413_35314 CHCF22 144947228

JN613413_53252 CHCF23 144965272/1609104181

JN613413_53278 CHCF24 144965298/1609103921

JN613413_53373 CHCF25 144965393/1609103011

JN613413_53494 CHCH26 144965514/1609101801

JN613413_53612 CHCF27 144965632/1609100621

JN613413_61641 CHCF28 144973813

JN613413_61768 CHCF29 144973940

JN613413_62196 CHCF30 144974368

MUC13_g.207 CHCF31 144986614

MUC13_g.791 CHCF32 144987198

MUC13_g.1248 CHCF33 144987655

MUC13_g.1345 CHCF34 144987751

MUC13_g.1412 CHCF35 144987819

Page 111: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

SUMMARY

110

Continued from previous page

SNPs NCBI

dbSNP ID

Position on

Sscrofa 10.2

MUC13_g.1414 CHCF36 144987821

MUC13_g.1945 CHCF37 144988352

MUC13_g.1974 CHCF38 144988381

MUC13_g.8951 CHCF39 144997010

MUC13_g.8981 CHCF40 144997040

MUC13_c.232 CHCF41 144997797

MUC13_g.15150 CHCF42 145003208

MUC13_g.15376 CHCF43 145003434

MUC13_g.15379 CHCF44 145003436

MUC13_g.15381 CHCF45 145003439

MUC13_c.1243 CHCF46 145019310

MUC13_c.1289 CHCF47 145019356

MUC13_c.1290 CHCF48 145019357

MUC13_c.1702 CHCF49 145019769

MUC13_c.1788 CHCF50 145019855

MUC13_c.1842 CHCF51 145019909

MUC13_c.1930 CHCF52 145019997

MUC13_c.1986 CHCF53 145020053

MUC13_c.2014 CHCF54 145020081

MUC13_c.2068 CHCF55 145020135

JN613413_107599 CHCF56 145021109

JN613413_107649 CHCF57 145021159

JN613413_115242 CHCF58 145028754

JN613413_115364 CHCF59 145028876

JN613413_115460 CHCF60 145028972

ITGB5_c.246 CHCF61 145064038

ITGB5_g.65464 CHCF62 145107986/1608950211

ITGB5_c.917 CHCF63 145108180/1608952151

ITGB5_g.115393 CHCF64 ---2

1 SNP maps twice on Sscrofa 10.2

2 SNP ITGB5_g.115393 does not map in reference sequence Sscrofa 10.2, this SNP is mapped in BAC

clone CH242-89F13, NCBI accession no. CU466522.

Page 112: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

SUMMARY

111

CURRICULUM VITAE

Antonio Rampoldi

Personal Information

Date of birth 26th March 1983

Place of birth Cantù, Italy

Nationality Italian

Citizen of Bregnano, Italy

Education and Training

Dates (from – to) 2002

Name and type of organisation

providing education and training

Liceo Scientifico “De Amicis”

Via Salita Camuzio 4 - 22063 Cantù - Italy

Title of qualification awarded Scientific high school diploma 92/100

Dates (from – to) 02/2005-06/2005

Name and address of employer Zootechnical Department, Biotechnical Faculty, University of

Ljubljana

3 GROBLJE - 1230 DOMŽALE - SLOVENIA

Type of business or sector ERASMUS Program

Occupation or position held Intern

Page 113: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

112

Dates (from – to) 22/02/2006

Name and type of organisation

providing education and training

UNIVERSITY OF MILAN

Via Festa del Perdono 7 – 20122 Milan - Italy

Title of qualification awarded Bachelor’s Degree in Veterinary biotechnology 110/110

Dates (from – to) 20/11/2007

Name and type of organisation

providing education and training

UNIVERSITY OF MILAN

Via Festa del Perdono 7 – 20122 Milan - Italy

Title of qualification awarded Master’s Degree in Veterinary biotechnology 110/110 with

distinction

Dates (from – to)

06/2008-07/2008

Name and address of employer Unité de Génomique et Physiologie de la Lactation

INRA

78352 Jouy-en-Josas Cedex, France

Type of business or sector Galileo Program

Occupation or position held Intern

Dates (from – to) 08/2008-07/2013

Name and address of employer Institut für Agrarwissenschaften (IAS)

ETH Zürich

8092 Zürich, Switzerland

Occupation or position held Doctoral student

Page 114: Research Collection · 2020. 3. 26. · - 6 - 2.2.2 Sampling of intestinal tissues 29 2.2.3 Preparation of enterocytes 30 2.2.4 Microscopic adhesion test 30 2.2.5 Positive and negative

113

Publications

Rampoldi A., Jacobsen M.J., Bertschinger H.U., Joller D., Bürgi E., Vögeli P., Andersson L., Archibald

A.L., Fredholm M., Jørgensen C.B., Neuenschwander S. (2011): The receptor locus for Escherichia coli

F4ab/F4ac in the pig maps distal to the MUC4-LMLN region. Mammalian Genome 22: 122-129.

Jacobsen M., Cirera S., Joller D., Esteso G., Kracht S.S., Edfors I., Bendixen C., Archibald A.L., Vogeli

P., Neuenschwander S., Bertschinger H.U., Rampoldi A., Andersson L., Fredholm M., Jørgensen C.B.

(2011): Characterisation of five candidate genes within the ETEC F4ab/F4ac candidate region in pigs.

BMC Research Notes 4: 225.

Abstracts

Rampoldi A., Bertschinger H.U., Bürgi E., Vögeli P., Joller D., Neuenschwander S. “Mapping of the

Escherichia coli F4ab/F4ac receptor locus on pig chromosome 13”. Pig Genome III Conference, 2-4

November, 2009, Hinxton, UK; poster.

Rampoldi A., Bertschinger H.U., Bürgi E., Vögeli P., Jacobsen M.J., Jørgensen C.B., Neuenschwander S.

“Refined mapping of the Escherichia coli F4ab/F4ac receptor locus (F4bcR) on pig chromosome 13”.

32nd Conference of the International Society of Animal Genetics, 26-30 July, 2010, Edinburgh, UK;

poster.

Neuenschwander S., Bruggmann R., Rampoldi A., Qi W., Aluri S., Schlapbach R., Vögeli P. “Detection

of porcine miRNA in various tissues by next generation sequencing”. 32nd Conference of the

International Society of Animal Genetics, 26-30 July, 2010, Edinburgh, UK; poster.

Rampoldi A., Jacobsen M.J., Bertschinger H.U., Joller D., Bürgi E., Vögeli P., Jørgensen C.B.,

Neuenschwander S. “Comparative and molecular approach to the identification of receptors for E. coli

with fimbriae F4ab/F4ac in the pig”. Schweizerische Vereinigung für Tierproduktion, 2011, Zollikofen,

Switzerland; poster.

Rampoldi A., Bertschinger H.U., Bürgi E., Vögeli P., Jørgensen C.B., Jacobsen M.J., Neuenschwander S.

“Exclusion of gene HEG1 as receptor locus for the E. coli F4ab/F4ac”. 33rd Conference of the

International Society for Animal Genetics, 15-20 July, 2012, Cairns, Australia; poster.

Rampoldi A., Bertschinger H.U., Bürgi E., Dolf G., Vögeli P., Neuenschwander S. “Inheritance

mechanisms of receptor(s) for Enterotoxigenic Escherichia coli fimbriae F4ad (F4adR) in the pig”.

Schweizerische Vereinigung für Tierproduktion, 2013, Posieux, Switzerland; poster.