Co-infection of Helicobacter pylori and Opisthorchis ...ï ñì ,1752'8&7,21 ñí...

32
1 Co-infection of Helicobacter pylori and Opisthorchis viverrini enhances the 1 severity of hepatobiliary abnormalities in hamsters 2 3 Rungtiwa Dangtakot 1, 2 , Somchai Pinlaor 3, 4 , Upsornsawan Itthitaetrakool 5 , Apisit Chaidee 3, 4 , 4 Chariya Chomvarin 6, 4 , Arunnee Sangka 2 , Chotechana Wilailuckana 2 , Porntip Pinlaor 2, 4* 5 6 1 Science Program in Medical Technology, Faculty of Associated Medical Sciences, Khon 7 Kaen University, Khon Kaen 40002, Thailand 8 2 Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of 9 Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand 10 3 Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 11 40002, Thailand 12 4 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen 13 40002, Thailand 14 5 Biomedical Science Program, Graduate School, Khon Kaen University, Khon Kaen, 15 Thailand 16 6 Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 17 40002, Thailand 18 19 * Corresponding author at: Department of Microbiology, Faculty of Associated Medical 20 Science, Khon Kaen University, Khon Kaen, Thailand. Tel.: +66 43 202086, fax: +66 43 21 202086. 22 E-mail address:[email protected] (P. Pinlaor). 23 24 IAI Accepted Manuscript Posted Online 30 January 2017 Infect. Immun. doi:10.1128/IAI.00009-17 Copyright © 2017 American Society for Microbiology. All Rights Reserved. on March 26, 2020 by guest http://iai.asm.org/ Downloaded from

Transcript of Co-infection of Helicobacter pylori and Opisthorchis ...ï ñì ,1752'8&7,21 ñí...

  • 1

    Co-infection of Helicobacter pylori and Opisthorchis viverrini enhances the 1

    severity of hepatobiliary abnormalities in hamsters 2

    3

    Rungtiwa Dangtakot1, 2, Somchai Pinlaor3, 4, Upsornsawan Itthitaetrakool5, Apisit Chaidee3, 4, 4

    Chariya Chomvarin6, 4, Arunnee Sangka2, Chotechana Wilailuckana 2, Porntip Pinlaor2, 4* 5

    6

    1 Science Program in Medical Technology, Faculty of Associated Medical Sciences, Khon 7

    Kaen University, Khon Kaen 40002, Thailand 8

    2 Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of 9

    Associated Medical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand 10

    3 Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 11

    40002, Thailand 12

    4 Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen 13

    40002, Thailand 14

    5 Biomedical Science Program, Graduate School, Khon Kaen University, Khon Kaen, 15

    Thailand 16

    6 Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 17

    40002, Thailand 18

    19

    * Corresponding author at: Department of Microbiology, Faculty of Associated Medical 20

    Science, Khon Kaen University, Khon Kaen, Thailand. Tel.: +66 43 202086, fax: +66 43 21

    202086. 22

    E-mail address:[email protected] (P. Pinlaor). 23

    24

    IAI Accepted Manuscript Posted Online 30 January 2017Infect. Immun. doi:10.1128/IAI.00009-17Copyright © 2017 American Society for Microbiology. All Rights Reserved.

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 2

    Running title: Co-infection of H. pylori and O. viverrini in liver. 25

    26

    ABSTRACT 27

    Persistent infection with Opisthorchis viverrini causes hepatobiliary abnormalities, 28

    predisposing infected individuals to cholangiocarcinoma (CCA). In addition, Helicobacter 29

    pylori is highly prevalent in most countries and is a possible risk factor for CCA; however, its 30

    role in enhancing hepatobiliary abnormality is unclear. Here, we investigated the effects of 31

    co-infection of H. pylori and O. viverrini on hepatobiliary abnormality. Hamsters were 32

    divided into four groups: (1) normal, (2) H. pylori-infected (HP), (3) O. viverrini-infected 33

    (OV), and (4) O. viverrini plus H. pylori-infected (OV+HP). At 6 months post-infection, PCR 34

    and immunohistochemistry were used to test for the presence of H. pylori in the stomach, 35

    gallbladder and liver. In the liver, H. pylori was detected in the order of OV+HP (5 of 8, 36

    62.5%), HP (2 of 5, 40%) and OV (2 of 8, 25%): H. pylori was not detected in normal 37

    (control) liver tissues. Co-infection induced the most severe hepatobiliary abnormalities, 38

    including periductal fibrosis, cholangitis and bile duct hyperplasia, leading to significantly 39

    decreased survival rate of experimental animals. The greatest thickness of periductal fibrosis 40

    was associated with a significant increase of fibrogenesis markers (expression of α-SMA and 41

    TGF-β). qRT-PCR revealed that the highest expression levels of pro-inflammatory cytokine 42

    genes (IL-1, IL-6 and TNF-α) were also observed in the OV+HP group. These results suggest 43

    that co-infection of H. pylori and O. viverrini increased severity of hepatobiliary 44

    abnormalities to a greater extent than either single infection. 45

    46

    Keywords: Helicobacter pylori, Opisthorchis viverrini, hepatobiliary disease, periductal 47

    fibrosis, cholangitis, hepatic inflammation 48

    49

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 3

    1. INTRODUCTION 50

    Infection with the human liver fluke, Opisthorchis viverrini, causes opisthorchiasis, 51

    which is endemic in the Greater Mekong Sub-region. In Thailand, approximately six million 52

    people are currently infected by O. viverrini and the highest prevalence rate is found in the 53

    northeastern regions (1, 2). This fluke is classified as a group 1 carcinogen by the 54

    International Agency for Research on Cancer because of its role in the development of 55

    cholangiocarcinoma (CCA) (3). Humans become infected after eating raw freshwater fish 56

    contaminated with the infective stage, the metacercaria. Newly excysted juvenile worms in 57

    the duodenum migrate into the hepatobiliary system and develop into adults within one 58

    month, promoting hepatobiliary abnormalities including hepatic dysplasia, bile duct 59

    hyperplasia, periductal fibrosis, and bile duct cancer (3). Fewer than 10 % of infected 60

    individuals in the community develop CCA (1, 2). 61

    Etiology of CCA is multifactorial. The importance of single risk factors, such as 62

    bacterial infection, can be hard to gauge. A meta-analysis revealed that Helicobacter species 63

    in the hepatobiliary system were associated with CCA globally, as well as in liver fluke-free 64

    areas (4). One member of the genus, Helicobacter pylori, has also been classified as a group I 65

    carcinogen and is common in the gastro-duodenal system (3). Recent reviews have reported 66

    that H. pylori infection is associated with several diseases outside the gastrointestinal tract, 67

    including in the hepatobiliary system (5, 6). The presence of H. pylori DNA in gallbladder 68

    and gastric mucosa has been strongly associated with clinicopathological features of chronic 69

    cholecystitis patients, a risk condition for CCA (7). More than fifty percent of the human 70

    population worldwide is colonized by H. pylori. Prevalences are particularly high in 71

    developing countries, including in the northeastern part of Thailand (8). Although H. pylori 72

    has been detected in various organs and products such as liver, bile, gallbladder and gallstone 73

    (9), its role in the development of hepatobiliary disease is unclear. 74

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 4

    In fluke-endemic areas in Thailand, H. pylori was found in 66.7% of the patients with 75

    CCA and 41.5% of the patients with cholelithiasis. The presence of H. pylori in the liver and 76

    gallbladder was associated with biliary inflammation and proliferation (10). Recently, H. 77

    pylori infection was found to be more prevalent in O. viverrini-infected hamsters than in 78

    normal hamsters, suggesting that the parasite in the bile duct is a reservoir of H. pylori and 79

    Helicobacter-like bacteria (11). Taken together, these lines of evidence support the 80

    hypothesis that H. pylori co-infection with O. viverrini synergistically increases the severity 81

    of hepatobiliary abnormalities. 82

    In order to test this hypothesis, the present study aims to clarify the effect of co-infection 83

    of H. pylori and O. viverrini on hepatobiliary abnormalities in a hamster model. The outcome 84

    of this study might help to explain the hepatobiliary changes in humans in fluke-endemic 85

    areas. It might also be useful for suggesting modifications to the therapeutic approaches for 86

    prevention and control of opisthorchiasis-associated CCA. 87

    88

    2. MATERIALS AND METHODS 89

    Bacterial strains 90

    The H. pylori strain DMST 20165 (cagA+, vacA+, ureA+), obtained from the 91

    National Institute of Health, Department of Medical Sciences, Ministry of Public Health, 92

    Thailand, was used in this study. Helicobacter pylori was grown on 10% sheep blood agar 93

    and incubated for 48-72 h at 37 °C under microaerobic conditions (10% CO2, 5% O2, and 94

    85% N2) using CampygenTM 3.5 L, Oxoid Ltd. Enrichment of bacterial growth on the plates 95

    was confirmed by biochemical tests including catalase, oxidase, urease and Gram staining. 96

    Next, H. pylori was subcultured in Brucella broth supplemented with 10% fetal bovine serum 97

    for 24 hour at 37°C under microaerobic condition in an incubator shaker and then was 98

    checked for urease, catalase and oxidase activity. After that, cells were suspended in 99

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 5

    phosphate-buffered saline (PBS) to an optical density at 600 nm of 1.000 (~109 colony-100

    forming units) (12) under microaerobic condition and were ready for inoculation by gastric 101

    intubation. 102

    Preparation of O. viverrini metacercariae 103

    The O. viverrini metacercariae were isolated from the naturally infected cyprinoid 104

    fishes by artificial pepsin digestion (0.25% Pepsin A, BDH, USA) as previously described 105

    elsewhere (13). Metacercariae of O. viverrini were identified under a stereo microscope. 106

    Animals 107

    Male Syrian golden hamsters (Mesocricetus auratus), 4-6 weeks old, were obtained 108

    from the Animal Unit, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand. 109

    These animals were used in this study because the anatomical structure of the hamster 110

    hepatobiliary system is similar to that of humans and it is a susceptible host for O. viverrini. 111

    Animals were housed under conventional conditions and given food and water ad libitum. 112

    The study protocol was reviewed and approved by the Animal Ethics Committee of Khon 113

    Kaen University (Reference No. 0514.1.75/21). The standard guidelines of Ethics of Animal 114

    Experimentation of the National Research Council of Thailand were followed in order to 115

    ensure the health and well-being of our study animals. All animals enrolled in the experiment 116

    were healthy and animal health status was monitored daily. Before use, cages were washed 117

    with Dettol, a liquid antiseptic and disinfectant, then with washing-up liquid (Sunlight, a 118

    cleaning product) and finally fully dried. Bedding was autoclaved before starting the 119

    experiment. Cages and bedding were changed 1-2 time a week. 120

    Experimental design 121

    Forty animals were divided into four groups: (1) normal controls (Normal, n = 5), (2) 122

    H. pylori-infected (HP, n = 10), (3) O. viverrini-infected (OV, n = 10) and (4) H. pylori plus 123

    O. viverrini (OV+HP, n = 15). Metacercariae of O. viverrini and H. pylori were administered 124

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 6

    to hamsters via gastric intubation. For the H. pylori-infected groups, hamsters were infected 125

    with 0.5 ml of H. pylori suspended in PBS (approximately 5x108 colony-forming units) 126

    within one hour of preparing the bacteria in microaerophilic conditions. For the O. viverrini-127

    infected groups, hamsters were infected with fifty fluke metacercariae in normal saline 128

    solution. Usually, H. pylori colonizes the stomach. To maximize the chances of the bacterium 129

    reaching the hepatobiliary system, H. pylori was administered to the hamsters one week 130

    before and one month after O. viverrini infection (by which time worms have matured and 131

    already produced inflammation-mediated bile-duct and liver injuries). 132

    Specimen collection 133

    At 6 months post-infection (p.i.), hamsters were sacrificed using sodium pentobarbital 134

    anesthesia. For DNA isolation, stomach, liver and gallbladder tissues were collected, snap-135

    frozen in liquid nitrogen and stored at -80 °C until used. For RNA isolation, livers were 136

    collected in TRIZOL™ reagent and stored at -80 °C until used. For histopathological and 137

    immunohistochemical studies, stomach and liver tissues were collected and fixed in 10% 138

    buffered formalin. Blood samples were taken from the heart, centrifuged at 3,000 rpm for 10 139

    min at 4°C, and serum was collected and divided into 400 µl aliquots and kept at -80°C until 140

    used for biochemical analyses. 141

    Histopathology and immunohistochemistry 142

    Hamster liver and stomach samples were fixed in neutral buffered 10% formalin, 143

    embedded in paraffin wax, sectioned at 5 µm, and stained with hematoxylin and eosin 144

    (H&E). The grade of cholangitis visible in liver sections was scored according to the 145

    infiltration of inflammatory cells as follows: grade 0, no cholangitis; grade 1, mild invasion 146

    of inflammatory cells around the bile duct; grade 2, severe invasion of inflammatory cells 147

    around the bile duct; grade 3, abscess formation in the liver (14). Deposition of collagen at 148

    sites of hepatic and periductal fibrosis was stained using picrosirius red kit (Polysciences, 149

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 7

    Inc., Warrington, PA, USA) according to the manufacturer’s instructions. Periductal fibrosis 150

    was graded under a bright-field microscope (Carl Zeiss, Jena, Germany) into five stages as 151

    follows: grade 0, no fibrosis; grade 1, mild fibrous expansion of some portal areas; grade 2, 152

    moderate fibrous expansion of most portal areas with short fibrous septa; grade 3, severe 153

    fibrous expansion of most portal areas with occasional portal-to-portal bridging; grade 4: 154

    more severe fibrous expansion of most portal areas with marked bridging (15). 155

    Presence of H. pylori in stomach and liver samples was identified by immunostaining 156

    with rabbit polyclonal anti-H. pylori (Abcam, Cambridge, MA, USA, ab7788; 1:10 dilution), 157

    counterstained with Mayer’s hematoxylin and examined using a bright-field microscope. 158

    Western blot analysis 159

    Hepatic fibrosis was also confirmed by the expression of α-smooth muscle actin. The 160

    concentration of protein extracted from a liver sample was determined using a Bio-Rad 161

    protein assay kit (Bio-Rad, Hercules, CA, USA). Twenty micrograms of protein was used for 162

    western blotting and stained with anti-α-smooth muscle actin (Abcam, ab5694, 1:1,000) and 163

    β-Tubulin (9F3) Rabbit mAb (Cell Signaling Technology, USA, #2128, 1:100) solution at 164

    4°C overnight. Immunostaining was detected using an ECL Prime Western Blotting 165

    Detection kit (GE Healthcare, Maidstone, UK). Relative band density was quantified using 166

    my Image Analysis v2.0 software (Life Technologies, Thermo Fisher Scientific, Carlsbad, 167

    CA, USA). 168

    DNA extraction 169

    Using a QiAamp Tissue kit (Qiagen, Germany), DNA was extracted from pieces of 170

    15-25 mg cut from frozen samples of stomach, liver and gallbladder tissues. DNA 171

    concentration was measured using a NanoDrop 2000 spectrophotometer (NanoDrop 172

    Technologies, Wilmington, DE, USA). The purity of DNA in individual samples was 173

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 8

    assessed by measuring absorbance at 260 and 280 nm: ratios of these values ranging from 1.7 174

    to 2.1 were considered acceptable. 175

    176

    PCR amplification 177

    Amplification of DNA extracted from the gastric, liver, and gallbladder tissues was 178

    done using Helicobacter genus-specific 16S rRNA primers. Identification of H. pylori was 179

    done by H. pylori ureA primers (primer sequences and PCR conditions in Table S1) (16). The 180

    primers were checked for specificity using BLAST searches at https://blast.ncbi.nlm.nih.gov. 181

    The 20 µl PCR reaction included 1x PCR buffer, 1 mM MgCl2, 0.3 mM dNTP, 0.25 µM each 182

    primer, 1U platinum Taq DNA polymerase, and 300 ng of DNA template. Negative controls 183

    without DNA template and positive controls with H. pylori DNA were run in parallel. The 184

    PCR reactions were run in a thermal cycler and an Expand high-fidelity PCR system (Bio 185

    Rad C1000TM Thermal cycler). The amplified PCR products were separated by 186

    electrophoresis on 1.5 % agarose gels along with 100 bp plus DNA ladder and visualized by 187

    UV light after staining by ethidium bromide. 188

    Gene expression study 189

    To determine mRNA expression of TNF-α, IL-1, IL-6 and TGF-β, total RNA was 190

    extracted from frozen hamster liver tissues using Trizol (Invitrogen, Carlsbad, CA, USA). 191

    The concentration of RNA was measured using a NanoDrop 2000 spectrophotometer 192

    (NanoDrop Technologies, Wilmington, DE, USA). The purity of RNA in individual samples 193

    was assessed by measuring absorbance at 260 and 280 nm: ratios of these values ranging 194

    from 1.99 to 2.1 were considered acceptable. Complementary DNA (cDNA) was synthesized 195

    using RevertAid reverse transcriptase (Thermo Scientific) and used for qRT-PCR. The 196

    specific primer pairs IL-1: 5´GCCCATCTTCTGTGACTCCT3´-F; 197

    5´TGGAGAACACCACTTGTTGG3´-R, IL-6: 5´GACTTCACAGAGGACACTAC3´-F; 198

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 9

    5´CACATAGTCATTGTCCATACAG3´-R, TNF-α: 5´GACGGGCTGTACCTGGTTTA3´-F; 199

    5´GAGTCGGTCACCTTTCTCCA3´-R, TGF-β: 5´ACATCGACTTTCGCAAGGAC3´-F; 200

    5´TGGTTGTAGAGGGCAAGGAC3´-R, and GAPDH: 201

    5´AGAAGACTGTGGATGGCCCC3´-F; 5´TGACCTTGCCCACAGCCTT3´-R were used 202

    for amplification of hamster genes. Relative mRNA expression was analyzed by 203

    LightCycler® 480 II using LightCycler® 480 SYBR Green I Master. All data were analyzed 204

    using LightCycler® 480 software and then processed using the 2−ΔΔCt method (17) relative to 205

    GAPDH mRNA. 206

    Biochemical analyses 207

    Serum aspartate transferase (AST), alanine transferase (ALT) and alkaline 208

    phosphatase (ALP), the indicators of liver and bile duct injury, were measured using an 209

    automated spectrophotometer (automate RA100) using a commercial kit (Thermo Trace Ltd., 210

    Melbourne, Australia). 211

    Statistical analysis 212

    To compare between two groups, non-parametric data were analyzed using Kruskal 213

    Wallis and Mann-Whitney U tests and parametric data were analyzed using analysis of 214

    variance. P values

  • 10

    hamsters infected with O. viverrini plus H. pylori. Thus, five hamsters of the normal group, 224

    five hamsters of the HP group, and eight hamsters in both the OV group and the OV+HP 225

    group were terminated at the designated time-point at 6 months. 226

    The decreased survival rate in the OV+HP group was statistically significant 227

    compared to the normal control group (P = 0.0105, Fig. 1), whereas either OV- or HP- 228

    treatment alone decreased survival rate, but not to a statistically significant level relative to 229

    controls. However, all normal hamsters were still alive at 6 months and did not show any 230

    clinical signs. 231

    Detection of H. pylori by PCR and immunohistochemistry 232

    The prevalence of H. pylori in all gastric, gallbladder and liver samples, as detected 233

    by PCR and immunohistochemistry, is summarized in Table 1. PCR analysis of liver samples 234

    revealed H. pylori DNA in 2 of 8 (25%), 2 of 5 (40%) and 4 of 8 (50%) of the OV, HP, and 235

    OV+HP groups, respectively: H. pylori DNA was not detected in any normal liver sample. 236

    Immunohistochemistry demonstrated H. pylori in liver sections from the HP group (1 237

    of 5, 20%) and from the OV+HP group (2 of 8, 25%), but not in the OV and normal groups 238

    (Fig. 2). 239

    Overall prevalences of H. pylori in gastric samples were 20%, 40%, 50% and 75% for 240

    normal, HP, OV and OV+HP groups, respectively. Corresponding results for liver samples 241

    were 25%, 40% and 62.5% for OV, HP and OV+HP groups, respectively. No H. pylori was 242

    found in the liver tissues of normal group. 243

    Histopathological finding 244

    Gastric lesions including ulcers, mucosal hyperplasia and gland proliferation were 245

    observed in one hamster of the HP group and one hamster of the OV+HP group. In contrast, 246

    such lesions were not observed in normal or OV groups (Fig. S1). 247

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 11

    Cholangitis was not observed in any hamster in the normal control group (Fig. 3a). 248

    Only one hamster of the HP group showed mild invasion of inflammatory cells around the 249

    bile duct (Fig. 3b). Around the bile ducts containing adult worms (OV and OV+HP groups), 250

    marked accumulations of inflammatory cells were evident (Fig. 3c and Fig. 3d). The most 251

    severe histopathological changes were observed in the OV+HP group. These changes 252

    included infiltration by inflammatory cells, thickening of periductal fibrosis, lymphoid 253

    follicles, and cholangitis. Cholangitis was observed in hamsters of the OV and OV+HP 254

    groups. The grade scores of cholangitis increased in the order HP group, OV group and 255

    OV+HP group (Table 2). Although the OV and OV+HP groups exhibited similar cholangitis 256

    grade scores, massive invasion of inflammatory cells around the bile duct, with abscess 257

    formation in the liver and bile duct hyperplasia, were more evident in the OV+HP group than 258

    in the OV group. 259

    Fibrosis in the hamster livers 260

    Liver collagen content was determined by picrosirius red staining (Fig. 4A), which 261

    facilitated scoring of fibrosis. The grade scores of fibrosis increased in the order HP group, 262

    OV group and OV+HP group (Table 2). All hamsters of the HP group exhibited mild fibrous 263

    expansion of some portal areas and parenchyma. For the OV group, all hamsters developed 264

    moderate to severe fibrous expansion of most portal areas with short fibrous septa or 265

    occasional portal-to-portal bridging. For the OV+HP group, two hamsters developed 266

    moderate fibrous expansion of most portal areas with short fibrous septa; four hamsters 267

    developed severe fibrous expansion of most portal areas with occasional portal-to-portal 268

    bridging, and two hamsters developed more severe fibrous expansion of most portal areas 269

    with marked bridging. Interestingly, the most severe fibrosis was observed in the OV+HP 270

    group, especially in individuals positive for H. pylori. No fibrosis was observed in the normal 271

    group. 272

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 12

    Expression of α-SMA in hamster livers 273

    To evaluate fibrosis, we investigated the expression level of α-SMA using western 274

    blot (Fig. 4B). In the normal and HP groups, α-SMA was hardly detected by western blotting. 275

    The greatest band intensity of α-SMA was observed in some of the OV group (2 of 8) and in 276

    the OV+HP group (5 of 8). Interestingly, relative intensities of α-SMA were significantly 277

    greater in the OV+HP group than in the OV group (p

  • 13

    where O. viverrini is endemic (8). Despite this, there have been no detailed studies of 298

    hepatobiliary abnormalities in the H. pylori-infected opisthorchiasis hamster model: the 299

    present study is the first to do this. As we know, in acute infection, O. viverrini releases 300

    antigens which activate host-parasite interactions resulting in inflammatory cell infiltration, 301

    excess of cytokines and free radical production, bile duct epithelial hyperplasia and injury of 302

    epithelial and hepatic cells. In chronic infection, myofibroblasts become activated and 303

    migrate into the damaged tissue and synthesize extracellular matrix (ECM) components 304

    leading to increased periductal fibrosis, which might be one possible risk condition for CCA 305

    development (2). 306

    Herein, we demonstrated the effect of co-infection of H. pylori and O. viverrini on 307

    histopathological changes in hamster liver. Co-infection promoted more severe hepatobiliary 308

    abnormalities than did either single infection. Pathological changes included cholangitis and 309

    periductal fibrosis, leading to decreased survival rate in hamsters. This was accompanied by 310

    up-regulated expression of genes encoding IL-1, IL-6, TNF-α, the proinflamatory cytokines, 311

    and TGF-β, the profibrotic cytokine, which showed the highest levels in the co-infected 312

    group. Cholangitis, periductal fibrosis and bile duct hyperplasia were also found to be the 313

    most severe in some hamster livers in the co-infected group. In addition, the expression of α-314

    SMA protein, an indicator of myofibroblasts, increased significantly in the group co-infected 315

    with both pathogens when compared to O. viverrini infection alone. 316

    Previously, we have sequentially reported that O. viverrini infection induces 317

    inflammation-mediated oxidative stress (18), leading to accumulated periductal fibrosis 318

    increasing with time (13) and that the fibrotic lesion plays an important role in CCA genesis 319

    in hamsters (19). In this study, periductal fibrosis was most prominent in hamsters co-320

    infected with H. pylori and O. viverrini, suggesting that H. pylori may also accelerate the 321

    fibrogenesis occurring during O. viverrini infection. 322

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 14

    The enhanced periductal fibrosis seen in the co-infected group was associated with increased 323

    expression of inflammatory genes such as IL-1, IL-6 and TNF-α, in addition to any IL-17 and 324

    IFN-gamma response that might occur due to H. pylori in gastric lesions (20). Upregulated 325

    expression of IL-6 has been noted previously in O. viverrini (21) and H. pylori single 326

    infections (22), which might synergistically induce inflammation-mediated liver injury 327

    through oxidative stress (18, 22). As a result of this, the lipopolysaccaride (LPS) of bacteria, 328

    including H. pylori, can induce TGF-β signaling through a toll-like receptor 4 (TLR4) (23), 329

    contributing to fibrogenesis. Synergistic induction of TGF-β induces activation of 330

    myofibroblasts, leading to synthesis of large amounts of extracellular component proteins to 331

    restore injured tissues. Finally, the excessive accumulation of ECM components promotes the 332

    formation of the most prominent periductal fibrosis during O. viverrini and H. pylori co-333

    infection. Relevantly, a combination of H. pylori and carbon tetrachloride treatment can 334

    enhance severity of hepatic fibrosis in animals (24). Also, in chronic hepatitis-C patients, co-335

    infection with H. pylori can enhance liver fibrosis and cirrhosis (25). 336

    It is notable that not all animals in the H. pylori-treated groups (HP and OV+HP 337

    groups) exhibited severe hepatobiliary abnormalities. Most of the individuals exhibiting 338

    abnormalities returned positive tests for H. pylori in the liver. In the HP group, two hamsters 339

    positive for H. pylori showed mild fibrous expansion of some portal areas and parenchyma, 340

    and one of them had highly elevated ALT and AST levels. In the OV+HP group, among five 341

    hamsters positive for H. pylori, severe periductal fibrosis with marked bridging was seen in 342

    four. Among those with severe hepatic lesions, three hamsters showed intense α-SMA protein 343

    expression and one hamster had highly increased serum levels of ALT and AST. In contrast, 344

    one hamster in the co-infection group was negative for H. pylori, but exhibited increased 345

    plasma ALT level and cholangitis, suggesting that other bacterial infections could not be 346

    excluded as an explanation. Similarly, two hamsters in the OV group with moderate to severe 347

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 15

    periductal fibrosis were negative for H. pylori and exhibited slightly increased levels of ALT 348

    and AST. 349

    The question arises as to whether other Helicobacter species might play a role in 350

    hepatobiliary disease. Hamsters have been found naturally infected in various tissues with 351

    several novel Helicobacter spp. (26-29) belonging to the H. bilis cluster, which is known to 352

    be involved in hepatobiliary lesions (26, 30). To support this, H. bilis could be detected in 353

    14.9% and 9.4% of patients with CCA and cholelithiasis, respectively, and was absent in the 354

    control group (10). On the other hand, H. pylori, a group 1 carcinogen, was detected in a 355

    higher prevalence (66.7% in the patients with CCA, 41.5% in patients with cholelithiasis and 356

    25.0% in control groups) than that of H. bilis (10). Therefore, we strongly believe that H. 357

    pylori is involved in the pathogenesis of hepatobiliary diseases rather than H. bilis in Thai 358

    patients (31). 359

    DNA of H. pylori has been detected in the liver tissue, bile, gallbladder and gallstones 360

    in patients with hepatobiliary diseases. It is not clear how H. pylori enters the hepatobiliary 361

    system. In mice inoculated orally with H. pylori, the stomach was the most common site 362

    (86%), but infection rate in the hepatobiliary system was only 40% (32). In the present study, 363

    in the HP group, H. pylori was detected in 40 % of liver samples, a prevalence similar to that 364

    in the gastric samples. In hamsters infected with O. viverrini alone, H. pylori was detected in 365

    50% of gastric tissues, but was less prevalent in the hepatobiliary system (25%) than in the 366

    HP group, suggesting that oral inoculation with H. pylori permits its transmission to the 367

    biliary system with or without the presence of flukes. There appears to be a background level 368

    of natural infection of gastric tissue (but not liver tissue) with H. pylori in our hamsters, 369

    which was similar to findings in a previously study (11, 16). Inoculation with additional H. 370

    pylori and/or the presence of worms was associated with presence of H. pylori in the biliary 371

    system. 372

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 16

    Moreover, our results showed that when H. pylori was orally inoculated with O. 373

    viverrini, detection rates of H. pylori in gastric and liver tissues increased to 75 % and 62.5%, 374

    respectively. The colonization rate of H. pylori was higher in the co-infected group than in 375

    either of the single-infection groups. Helicobacter pylori may reach the liver in three possible 376

    ways: i) by causing gastric injury facilitating transport to the liver via the blood stream (33), 377

    ii) when inoculated with O. viverrini, H. pylori may be carried into the hepatobiliary system 378

    directly by the worms (11), or iii) physical obstruction of the bile ducts by O. viverrini 379

    infection (34) may lead to increased pressure and low pH within the biliary tract, which might 380

    permit influx of H. pylori from the gastrointestinal tract into the hepatobiliary system. 381

    In addition, H. pylori was detected by PCR but not by immunostaining in two 382

    hamsters inoculated with O. viverrini alone. This may have represented natural infection. 383

    Alternatively, H. pylori DNA in the stomach may have been passively carried to the 384

    hepatobiliary system (35). In some individual hamsters inoculated with H. pylori, the 385

    bacterium could be detected in the liver sample using PCR or immunohistochemical staining, 386

    but not in the gastric sample. This might be due to low numbers of bacteria colonizing the 387

    gastric site. Prevalences of H. pylori were similar in the gall bladder in the OV and OV+HP 388

    groups. This may be due to a limitation in this study that we couldn’t collect bile because of a 389

    significant decrease in bile production from as early as one month post-infection. Moreover, 390

    the status of our hamsters with regard to Helicobacter infection was not determined before 391

    starting the experiment. Nor did we determine the genetic profile of H. pylori (e.g. with 392

    respect to cagA and vacA genes) in the samples. However, we did inoculate our experimental 393

    animals with a virulent H. pylori strain (DMST20165), which is cagA+ and vacA+. These 394

    virulence genes are likely to enhance severity of hepatobiliary diseases (36) and peptic ulcers 395

    (37). We believe that a virulent strain of H. pylori is more likely to accelerate induction of 396

    hepatobiliary diseases in addition to O. viverrini infection alone. 397

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 17

    In summary, this study showed that H. pylori orally inoculated could cause not only 398

    gastric lesions but also could reach the hepatobiliary system, enhancing severity of 399

    hepatobiliary abnormalities such as periductal fibrosis and cholangitis, leading to 400

    significantly decreased survival rates in experimental opisthorchiasis. Longitudinal studies 401

    using a larger number of animals and extensive studies of H. pylori virulence gene profiles 402

    should be performed for risk assessment of opisthorchiasis-associated CCA to confirm this 403

    finding. This provides greater understanding of the effect of H. pylori not only in 404

    experimental opisthorchiasis, but also in co-infection of H. pylori with other helminth-related 405

    hepatobiliary diseases. In addition, it might be useful when considering relevant therapeutic 406

    approaches in human opisthorchiasis-associated CCA. 407

    408

    AUTHOR CONTRIBUTIONS 409

    PP, SP, CC, AS and CW contributed to the experimental design of the study. RD, UI 410

    and AC performed the experiments, data analysis, and drafted the manuscript. PP and SP read 411

    and corrected the manuscript. 412

    413

    COMPETING INTERESTS: the authors have no competing interests. 414

    415

    ACKNOWLEDGEMENTS 416

    This work was supported by a grant of The Thailand Research Fund (TRG5680032), 417

    and Khon Kaen University Research Fund (KKU590302). Miss Rungtiwa Dangtakot was 418

    supported by Centre for Research and Development of Medical Diagnostic Laboratories, 419

    Faculty of Associated Medical Sciences, KhonKaen University. We thank The National 420

    Institute of Health, Department of Medical Sciences, Ministry of Public Health, Thailand, for 421

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 18

    providing Helicobacter pylori strain DMST20165. We also thank Prof. David Blair at the 422

    publication clinic for his advice and English presentation. 423

    424

    REFERNCES 425

    1. Sithithaworn P, Yongvanit P, Duenngai K, Kiatsopit N, Pairojkul C. 2014. Roles of 426

    liver fluke infection as risk factor for cholangiocarcinoma. J. Hepatobiliary Pancreat. Sci. 427

    21:301-308. 428

    2. Sripa B, Brindley PJ, Mulvenna J, Laha T, Smout MJ, Mairiang E, Bethony JM, 429

    Loukas A. 2012. The tumorigenic liver fluke Opisthorchis viverrini--multiple pathways 430

    to cancer. Trends Parasitol. 28:395-407. 431

    3. IARC. 2012. Biological agents. A review of human carcinogens. IARC Monogr. Eval. 432

    Carcinog. Risks Hum. 100(B):1-441. 433

    4. Xiao M, Gao Y, Wang Y. 2014. Helicobacter species infection may be associated with 434

    cholangiocarcinoma: a meta-analysis. Int. J. Clin. Pract. 68:262-270. 435

    5. Franceschi F, Zuccala G, Roccarina D, Gasbarrini A. 2014. Clinical effects of 436

    Helicobacter pylori outside the stomach. Nat. Rev. Gastroenterol. Hepatol. 11:234-242. 437

    6. Roubaud Baudron C, Franceschi F, Salles N, Gasbarrini A. 2013. Extragastric 438

    diseases and Helicobacter pylori. Helicobacter 18 Suppl 1:44-51. 439

    7. Zhou D, Guan WB, Wang JD, Zhang Y, Gong W, Quan ZW. 2013. A comparative 440

    study of clinicopathological features between chronic cholecystitis patients with and 441

    without Helicobacter pylori infection in gallbladder mucosa. PLoS One 8:e70265. 442

    8. Uchida T, Miftahussurur M, Pittayanon R, Vilaichone RK, Wisedopas N, 443

    Ratanachu-Ek T, Kishida T, Moriyama M, Yamaoka Y, Mahachai V. 2015. 444

    Helicobacter pylori Infection in Thailand: A Nationwide Study of the CagA Phenotype. 445

    PLoS One 10:e0136775. 446

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 19

    9. Lee JW, Lee DH, Lee JI, Jeong S, Kwon KS, Kim HG, Shin YW, Kim YS, Choi 447

    MS, Song SY. 2010. Identification of Helicobacter pylori in Gallstone, Bile, and Other 448

    Hepatobiliary Tissues of Patients with Cholecystitis. Gut Liver 4:60-67. 449

    10. Boonyanugomol W, Chomvarin C, Sripa B, Bhudhisawasdi V, Khuntikeo N, 450

    Hahnvajanawong C, Chamsuwan A. 2012. Helicobacter pylori in Thai patients with 451

    cholangiocarcinoma and its association with biliary inflammation and proliferation. HPB 452

    (Oxford) 14:177-184. 453

    11. Deenonpoe R, Chomvarin C, Pairojkul C, Chamgramol Y, Loukas A, Brindley PJ, 454

    Sripa B. 2015. The carcinogenic liver fluke Opisthorchis viverrini is a reservoir for 455

    species of Helicobacter. Asian Pac. J. Cancer Prev. 16:1751-1758. 456

    12. Lee CW, Rao VP, Rogers AB, Ge Z, Erdman SE, Whary MT, Fox JG. 2007. Wild-457

    type and interleukin-10-deficient regulatory T cells reduce effector T-cell-mediated 458

    gastroduodenitis in Rag2-/- mice, but only wild-type regulatory T cells suppress 459

    Helicobacter pylori gastritis. Infect. Immun. 75:2699-2707. 460

    13. Prakobwong S, Pinlaor S, Yongvanit P, Sithithaworn P, Pairojkul C, Hiraku Y. 461

    2009. Time profiles of the expression of metalloproteinases, tissue inhibitors of 462

    metalloproteases, cytokines and collagens in hamsters infected with Opisthorchis 463

    viverrini with special reference to peribiliary fibrosis and liver injury. Int. J. Parasitol. 464

    39:825-835. 465

    14. Kitajima T, Tajima Y, Matsuzaki S, Kuroki T, Fukuda K, Kanematsu T. 2003. 466

    Acceleration of spontaneous biliary carcinogenesis in hamsters by bilioenterostomy. 467

    Carcinogenesis 24:133-137. 468

    15. Goodman ZD. 2007. Grading and staging systems for inflammation and fibrosis in 469

    chronic liver diseases. J. Hepatol. 47:598-607. 470

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 20

    16. Itthitaetrakool U, Pinlaor P, Pinlaor S, Chomvarin C, Dangtakot R, Chaidee A, 471

    Wilailuckana C, Sangka A, Lulitanond A, Yongvanit P. 2016. Chronic Opisthorchis 472

    viverrini Infection Changes the Liver Microbiome and Promotes Helicobacter Growth. 473

    PLoS One 11:e0165798. 474

    17. Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data using real-475

    time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25:402-408. 476

    18. Pinlaor S, Hiraku Y, Ma N, Yongvanit P, Semba R, Oikawa S, Murata M, Sripa B, 477

    Sithithaworn P, Kawanishi S. 2004. Mechanism of NO-mediated oxidative and 478

    nitrative DNA damage in hamsters infected with Opisthorchis viverrini: a model of 479

    inflammation-mediated carcinogenesis. Nitric Oxide 11:175-183. 480

    19. Prakobwong S, Yongvanit P, Hiraku Y, Pairojkul C, Sithithaworn P, Pinlaor P, 481

    Pinlaor S. 2010. Involvement of MMP-9 in peribiliary fibrosis and 482

    cholangiocarcinogenesis via Rac1-dependent DNA damage in a hamster model. Int. J. 483

    Cancer 127:2576-2587. 484

    20. Bhuiyan TR, Islam MM, Uddin T, Chowdhury MI, Janzon A, Adamsson J, Lundin 485

    SB, Qadri F, Lundgren A. 2014. Th1 and Th17 responses to Helicobacter pylori in 486

    Bangladeshi infants, children and adults. PLoS One 9:e93943. 487

    21. Sripa B, Mairiang E, Thinkhamrop B, Laha T, Kaewkes S, Sithithaworn P, 488

    Tessana S, Loukas A, Brindley PJ, Bethony JM. 2009. Advanced periductal fibrosis 489

    from infection with the carcinogenic human liver fluke Opisthorchis viverrini correlates 490

    with elevated levels of interleukin-6. Hepatology 50:1273-1281. 491

    22. Piao JY, Lee HG, Kim SJ, Kim DH, Han HJ, Ngo HK, Park SA, Woo JH, Lee JS, 492

    Na HK, Cha YN, Surh YJ. 2016. Helicobacter pylori Activates IL-6-STAT3 Signaling 493

    in Human Gastric Cancer Cells: Potential Roles for Reactive Oxygen Species. 494

    Helicobacter 21:405-416. 495

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 21

    23. Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA, Schwabe 496

    RF. 2007. TLR4 enhances TGF-β signaling and hepatic fibrosis. Nat. Med. 13:1324-497

    1332. 498

    24. Goo MJ, Ki MR, Lee HR, Yang HJ, Yuan DW, Hong IH, Park JK, Hong KS, Han 499

    JY, Hwang OK, Kim DH, Do SH, Cohn RD, Jeong KS. 2009. Helicobacter pylori 500

    promotes hepatic fibrosis in the animal model. Lab. Invest. 89:1291-1303. 501

    25. Sakr SA, Badrah GA, Sheir RA. 2013. Histological and histochemical alterations in 502

    liver of chronic hepatitis C patients with Helicobacter pylori infection. Biomed 503

    Pharmacother. 67:367-374. 504

    26. Fox JG, Shen Z, Muthupalani S, Rogers AR, Kirchain SM, Dewhirst FE. 2009. 505

    Chronic hepatitis, hepatic dysplasia, fibrosis, and biliary hyperplasia in hamsters 506

    naturally infected with a novel Helicobacter classified in the H. bilis cluster. J. Clin. 507

    Microbiol. 47:3673-3681. 508

    27. Patterson MM, Schrenzel MD, Feng Y, Xu S, Dewhirst FE, Paster BJ, Thibodeau 509

    SA, Versalovic J, Fox JG. 2000. Helicobacter aurati sp. nov., a urease-positive 510

    Helicobacter species cultured from gastrointestinal tissues of Syrian hamsters. J. Clin. 511

    Microbiol. 38:3722-3728. 512

    28. Simmons JH, Riley LK, Besch-Williford CL, Franklin CL. 2000. Helicobacter 513

    mesocricetorum sp. nov., A novel Helicobacter isolated from the feces of Syrian 514

    hamsters. J. Clin. Microbiol. 38:1811-1817. 515

    29. Zenner L. 1999. Pathology, diagnosis and epidemiology of the rodent Helicobacter 516

    infection. Comp. Immunol. Microbiol. Infect. Dis. 22:41-61. 517

    30. Woods SE, Ek C, Shen Z, Feng Y, Ge Z, Muthupalani S, Whary MT, Fox JG. 2016. 518

    Male Syrian Hamsters Experimentally Infected with Helicobacter spp. of the H. bilis 519

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 22

    Cluster Develop MALT-Associated Gastrointestinal Lymphomas. Helicobacter 21:201-520

    217. 521

    31. Matsukura N, Yokomuro S, Yamada S, Tajiri T, Sundo T, Hadama T, Kamiya S, 522

    Naito Z, Fox JG. 2002. Association between Helicobacter bilis in bile and biliary tract 523

    malignancies: H. bilis in bile from Japanese and Thai patients with benign and malignant 524

    diseases in the biliary tract. Jpn. J. Cancer Res. 93:842-847. 525

    32. Huang Y, Tian XF, Fan XG, Fu CY, Zhu C. 2009. The pathological effect of 526

    Helicobacter pylori infection on liver tissues in mice. Clin. Microbiol. Infect. 15:843-527

    849. 528

    33. Huang Y, Fan XG, Tang ZS, Liu L, Tian XF, Li N. 2006. Detection of Helicobacter 529

    pylori DNA in peripheral blood from patients with peptic ulcer or gastritis. APMIS 530

    114:851-856. 531

    34. Kimura Y, Takada T, Kawarada Y, Nimura Y, Hirata K, Sekimoto M, Yoshida M, 532

    Mayumi T, Wada K, Miura F, Yasuda H, Yamashita Y, Nagino M, Hirota M, 533

    Tanaka A, Tsuyuguchi T, Strasberg SM, Gadacz TR. 2007. Definitions, 534

    pathophysiology, and epidemiology of acute cholangitis and cholecystitis: Tokyo 535

    Guidelines. J. Hepatobiliary Pancreat. Surg. 14:15-26. 536

    35. Shukla HS, Tewari M. 2012. Discovery of Helicobacter pylori in gallbladder. Indian J. 537

    Gastroenterol. 31:55-56. 538

    36. Boonyanugomol W, Chomvarin C, Sripa B, Chau-In S, Pugkhem A, Namwat W, 539

    Wongboot W, Khampoosa B. 2012. Molecular analysis of Helicobacter pylori virulent-540

    associated genes in hepatobiliary patients. HPB (Oxford) 14:754-763. 541

    37. van Doorn LJ, Figueiredo C, Sanna R, Plaisier A, Schneeberger P, de Boer W, 542

    Quint W. 1998. Clinical relevance of the cagA, vacA, and iceA status of Helicobacter 543

    pylori. Gastroenterol. 115:58-66. 544

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 23

    545

    FIGURES AND LEGENDS 546

    FIG. 1. Survival rate of animals in the H. pylori-infected (HP) group, O. viverrini-infected 547

    (OV) group and in the group co-infected with O. viverrini and H. pylori (OV+HP), compared 548

    to normal hamsters. 549

    550

    FIG. 2. Representative images of immunohistochemistry using antibody against H. pylori 551

    demonstrating the bacterium in gastric and liver tissues. No colonization of H. pylori in 552

    gastric (a, e) and in liver tissues (b, f) was found in control hamsters (Normal) or in O. 553

    viverrini-infected hamsters (OV). Colonization of H. pylori was observed in a gastric pit (c 554

    and d) and in the liver tissues (g, h) of H. pylori-infected hamsters (HP) and in H. pylori plus 555

    O. viverrini-infected hamsters (OV+HP). Arrows indicate positive signal. Scale bar, 10 μm. 556

    557

    FIG. 3. Representative images of hepatobiliary abnormalities in H&E-stained sections. Panel 558

    (a), normal liver and hepatocytes; (b), liver tissue after H. pylori infection; (c), (e), (g), liver 559

    tissues of O. viverrini-infected hamsters. Panels (c) and (e) show inflammatory cell 560

    infiltration around the bile duct with and without O. viverrini inside. Panel (g) shows 561

    inflammatory cell infiltration in liver tissues. Panels (d), (f), (h), liver tissues of O. viverrini 562

    plus H. pylori-infected hamsters; panel (d) shows inflammatory cell infiltration around the 563

    bile duct within which is an adult O. viverrini. Panel (f) shows massive inflammatory cell 564

    infiltration and bile duct hyperplasia in an O. viverrini plus H. pylori-infected hamster. Panel 565

    (h) shows liver abscess formation. Scale bar 100 µm. 566

    567

    FIG. 4. Representative images of fibrosis in liver tissue visualized using picrosirius red 568

    staining (A) and expression of α-SMA protein in liver tissues (B) and its abundance relative 569

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 24

    to β-tubulin (C). Twenty six samples were run in two gels at the same time. Pooled sample 570

    was included in both gels. Each group of experiment was separated into different image. 571

    Western blot image of OV group was obtained from two different gels as indicated by thin 572

    line (B, the third set of images-between OV3 and OV4). *P

  • 25

    TABLE 1. The prevalence of H. pylori in gastric, liver and gallbladder samples, as detected 582

    by PCR and immunohistochemistry. 583

    584

    585

    586

    587

    588

    589

    590

    591

    592

    593

    594

    595

    596

    597

    598

    ND = Not determined 599

    600

    Group Specimen H. pylori

    positive by

    PCR

    H. pylori

    positive by

    IHC

    Total H.

    pylori

    positive

    Normal

    (n = 5)

    Stomach 20%(1/5) 0%(0/5) 20%(1/5)

    Liver 0%(0/5) 0%(0/5) 0%(0/5)

    Gallbladder 0%(0/5) ND 0%(0/5)

    HP

    (n = 5)

    Stomach 40%(2/5) 20%(1/5) 40%(2/5)

    Liver 40%(2/5) 20%(1/5) 40%(2/5)

    Gallbladder 0%(0/5) ND 0%(0/5)

    OV

    (n = 8)

    Stomach 50%(4/8) 0%(0/8) 50%(4/8)

    Liver 25%(2/8) 0%(0/8) 25%(2/8)

    Gallbladder 12.5%(1/8) ND 12.5%(1/8)

    OV+HP

    (n = 8)

    Stomach 62.5%(5/8) 62.5%(5/8) 75%(6/8)

    Liver 50%(4/8) 25%(2/8) 62.5%(5/8)

    Gallbladder 12.5%(1/8) ND 12.5%(1/8)

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 26

    TABLE 2. Effects of co-infection with H. pylori and O. viverrini on fibrosis grade, 601

    cholangitis grade, and serum biochemical factors in experimental animals. 602

    603

    604

    605

    Experimental

    group

    Fibrosis

    grade

    (Mean±SD)

    Cholangitis

    grade

    (Mean±SD)

    ALT (U/L)

    (Mean±SD)

    AST (U/L)

    (Mean±SD)

    ALP (U/L)

    (Mean±SD)

    Normal 0.17

    ±0.19

    0.00

    ±0.00

    41.60

    ±2.07

    47.20

    ±4.32

    55.00

    ±16.54

    HP 1.07

    ±0.36*

    0.20

    ±0.44

    52.40

    ±39.16

    60.80

    ±28.93

    54.40

    ±11.65

    OV 2.79

    ±0.43*,†

    2.18

    ±0.26*

    57.63

    ±17.32

    59.75

    ±12.22

    58.75

    ±3.88

    OV+HP 3.29

    ±0.60*,†

    2.56

    ±0.50*,†

    74.13

    ±46.08

    68.75

    ±17.96

    55.63

    ±18.63

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • 27

    606

    607

    on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/

  • on March 26, 2020 by guest

    http://iai.asm.org/

    Dow

    nloaded from

    http://iai.asm.org/