helicobacter pylori and epstein-barr virus coinfection stimulates … · 2020. 11. 19. · 47...

40
1 Title: Helicobacter pylori and Epstein-Barr virus coinfection stimulates the 1 aggressiveness in gastric cancer through the regulation of gankyrin 2 3 4 Authors: Dharmendra Kashyap a , Budhadev Baral a# , Nidhi Varshney a# , Anil Kumar Singh b , 5 Hem Chandra Jha a* 6 7 8 Affiliations: a = Discipline of Biosciences and Biomedical Engineering, Indian Institute of 9 Technology Indore, India 10 b= Biological Science and Technology Division, CSIR-North East Institute of Science and 11 Technology, India 12 13 14 15 16 # Equal contributors 17 * Corresponding author 18 19 20 21 22 Author for correspondence: Dr. Hem Chandra Jha, Infection Bio-engineering group, 23 Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology 24 Indore. Madhya Pradesh, PIN-453552, 0732-4306581 25 E-mail: [email protected] 26 Phone: +91-9971653189 27 Running title: Interplay between H. pylori and EBV through gankyrin 28 29 30 . CC-BY-NC-ND 4.0 International license perpetuity. It is made available under a preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in The copyright holder for this this version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807 doi: bioRxiv preprint

Upload: others

Post on 27-Jan-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

  • 1

    Title: Helicobacter pylori and Epstein-Barr virus coinfection stimulates the 1

    aggressiveness in gastric cancer through the regulation of gankyrin 2

    3

    4

    Authors: Dharmendra Kashyapa, Budhadev Baral

    a#, Nidhi Varshney

    a#, Anil Kumar Singh

    b, 5

    Hem Chandra Jhaa*

    6

    7

    8

    Affiliations: a = Discipline of Biosciences and Biomedical Engineering, Indian Institute of 9

    Technology Indore, India 10

    b= Biological Science and Technology Division, CSIR-North East Institute of Science and 11

    Technology, India 12

    13

    14

    15

    16

    # Equal contributors 17

    * Corresponding author 18

    19

    20

    21

    22

    Author for correspondence: Dr. Hem Chandra Jha, Infection Bio-engineering group, 23

    Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology 24

    Indore. Madhya Pradesh, PIN-453552, 0732-4306581 25

    E-mail: [email protected] 26

    Phone: +91-9971653189 27

    Running title: Interplay between H. pylori and EBV through gankyrin 28

    29

    30

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 2

    Abstract 31

    Persistent coinfection of Helicobacter pylori (H. pylori) and Epstein-Barr virus (EBV) 32

    promotes aggressive gastric carcinoma. The molecular mechanisms underlying the 33

    aggressiveness in H. pylori and EBV coinfected gastric cancer is not well characterized. In 34

    the current study, we investigated the molecular mechanism involved in the cooperation of H. 35

    pylori and EBV-driven proliferation of gastric epithelial cells. Results showed that the 36

    coinfections are significantly more advantageous to the pathogens to create a 37

    microenvironment that favors the higher pathogen-associated gene expression. The EBV 38

    latent genes EBNA1 and EBNA3C are highly overexpressed in the coinfections compared to 39

    individual EBV infection at different time points (12 and 24 hrs). The H. pylori-associated 40

    genes 16s rRNA, CagA, and BabA has also been highly overexpressed in coinfections 41

    compared to H. pylori alone. Gankyrin is a small protein of 25 KDa involved in multiple 42

    biological and physiological processes. The upregulation of gankyrin modulates the various 43

    cell signaling pathways, leading to oncogenesis. The gankyrin shows a similar expression 44

    pattern as EBNA3C at both transcript and protein levels, suggesting a possible correlation. 45

    Further EBV and H. pylori create microenvironments that induce cell transformation and 46

    oncogenesis by dysregulation of the cell-cycle regulator, GC marker, cell migration, DNA 47

    response, and antiapoptotic genes in infected gastric epithelial cells by enhancing the 48

    expression of gankyrin. Our study provides new insights into the molecular mechanism 49

    where the interplay between two oncogenic agents (H. pylori and EBV) leads to the enhanced 50

    carcinogenic activity of gastric epithelial cells through overexpression of oncoprotein 51

    gankyrin. 52

    Importance 53

    In the present study, we have evaluated the synergistic effect of EBV and H. pylori infection 54

    on gastric epithelial cells in various coinfection models. These coinfection models depict the 55

    first exposures of gastric epithelial cells with EBV and then the H. pylori. While other 56

    coinfection models narrated the first exposures of H. pylori followed by the infection of EBV. 57

    This led to an enhanced oncogenic phenotype in gastric epithelial cells. We determined the 58

    coinfection of EBV and H. pylori enhanced the expression of oncogenic protein gankyrin. 59

    The interplay between EBV and H. pylori promotes the oncogenic properties of AGS cells 60

    through the newly discovered oncoprotein gankyrin. EBV and H. pylori mediated 61

    upregulation of gankyrin further dysregulates various cancer-associated hallmarks of genes 62

    such as cell-migratory, gastric cancer marker, tumor suppressor, DNA damage response, and 63

    proapoptotic genes. 64

    Keywords: Helicobacter pylori, Epstein Barr Virus, Gankyrin, Gastric cancer, Coinfection 65

    Introduction 66

    Gastric cancer (GC) is the worlds' fourth leading cause of cancer-related deaths in both males 67

    and females (1). Helicobacter pylori (H. pylori) and Epstein-Barr virus (EBV) are group1 68

    carcinogens that potentially contribute to the development of GC (2). While doing so it can 69

    alter gastric physiology and immunology (3). The prevalence of H. pylori in about half of the 70

    global populations but it causes GC only in 3% of infected individuals (4, 5). The 71

    discrepancy in H. pylori infection and GC might be influenced by its strain variability such as 72

    expression of CagA, VacA, and BabA (6, 7, 8). EBV is the second most prominent cancer-73

    associated pathogen involved in several types of lymphoid and epithelial cancers, including 74

    GC (9). Latent and lytic genes expression associated with EBV such as EBNA-1, EBNA-2, 75

    EBAN-3C, and BZLF-1 provokes the disruption of the host genes (9). The viral proteins also 76

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 3

    dysregulated the cell cycle, inflammation, angiogenesis, and tumor suppressor genes by hyper 77

    methylation activities (10, 11). Earlier studies have shown the ability of EBV for oncogenic 78

    transformation of primary gastric epithelial cells (12, 13). Thus, it is not only considered as a 79

    passive carrier but also as an active oncogenic virus contributing to early events in the 80

    development of GC (2). Lone infection of H. pylori or EBV is less severe in comparison to 81

    coinfection and takes more time for the development of GC (2). 82

    Gankyrin or PSMD-10 is a recently discovered small protein of 25 KDa known to be 83

    overexpressed in various cancers (14). It is a novel oncogenic protein, involved in various 84

    cellular processes such as cell cycle progression, apoptosis, and tumorigenesis (15). Gankyrin 85

    negatively regulates the protein retinoblastoma (pRb) and tumor suppressor protein-53 86

    (p53/TP53) to execute its oncogenic functions (16,17). Gankyrin was downregulated in GC 87

    which leads to increased cell chemosensitivity to 5-fluorouracil and cisplatin was attained by 88

    regulating cell cycle-related proteins, such as cyclin D1, cyclin E, and by activating the 89

    PI3K/AKT pathway (18). 90

    The mutualistic relationship which endures within the microbial niche could modulate host 91

    gene expression, thus impact pathophysiology. We determined the molecular mechanism to 92

    elucidate the unexplored strategy, which involves the participation of H. pylori in EBV-93

    driven proliferation of gastric epithelial cells. We have developed five different infection 94

    model including one uninfected control, using AGS human gastric epithelial cells, which is 95

    an excellent system mimicking the human gastric epithelium. We elucidated that EBV and H. 96

    pylori increased the gankyrin expression which led to the proliferation of infected cells in our 97

    studied system. Furthermore, we determined the status of cell migratory, cell-cycle 98

    regulatory, DNA repair, apoptosis, and tumor suppressor genes in coinfection models which 99

    can be directly linked with aggressive carcinogenesis. 100

    Results: 101

    Coinfection of H. pylori and EBV synergistically regulates their genes expression in the 102

    gastric epithelial cell after infection establishment 103

    In this study, we evaluated the synergistic effect of H. pylori and EBV on gastric epithelial 104

    cells. To understand the synergistic effect of these two pathogens we have developed the in-105

    vitro coinfection models by using adenogastric carcinoma cell line (AGS). This cell line 106

    mimics the gastric epithelial cells and provides the same microenvironment for the growth of 107

    these two pathogens as the in-vivo conditions except pH (1.5-3.5). Before incubation of H. 108

    pylori with AGS cells we have checked its morphology through the Gram staining, as 109

    predicted we have observed the regular spiral shape of this bacterium (supplementary fig.1). 110

    We have also observed the interaction between the H. pylori and EBV and how their 111

    synergistic interplay affects the pathogenic gene expression of these pathogens and 112

    downstream host cellular physiology. Our results showed that the infection-III and Infection-113

    IV are significantly more advantageous to the pathogens to create a microenvironment that 114

    favors the higher pathogens associated gene expression as compared to individual infection at 115

    a time (Fig2). The expression of pathogens associated with carcinogenic genes is multiple 116

    folds higher in coinfection models in comparison to individual infection (Fig2 A&B). Both 117

    H. pylori and EBV synergistically provided a suitable milieu for the growth of each other. 118

    Subsequently, we have followed all the above models to understand the pathophysiology of 119

    these two pathogens in GC progression and severity and how these two pathogens mutually 120

    favor the infectivity of each other. Thus, after coinfection, we have collected the cells at 121

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 4

    various time points (12, 24, and 48 hrs) and checked the profiles of the pathogenic and host 122

    gene expression. 123

    After infection, we performed the qRT-PCR for the EBV-associated latent and lytic genes 124

    such as EBNA1, EBNA3C, and BZLF1 respectively. Our results showed that the expression 125

    of EBNA1 transcript is significantly higher in infection-III and IV at time point 12 (p=0.0027 126

    and 0.054), 24 (p=0.0024 and 0.0021), and 48 (p=0.025 and 0.0021) hrs as compared to the 127

    infection-II and I Fig2A (I). In this experiment, we have selected one uninfected negative 128

    control. This mock confirmed to us that there was no cross-contamination of these two 129

    pathogens and the mock was not compromised. Similarly, the nuclear antigen EBNA 3C 130

    expression was significantly higher in infection-IV at 12hrs post-infection (p=0.0450 and 131

    0.0022) as compared to infection-II and I whereas in the case of 24hrs of coinfected samples 132

    the expression of EBNA3C was higher in infection III as compared to infection-II and I 133

    showing the interplay between the EBV and H. pylori. However, the expression is 134

    significantly dropped at 48hrs post-infection Fig2A (II). 135

    Furthermore, the expression of BZLF-1 transcript is higher (~100-150 folds) in infection-II, 136

    while decreasing in infection-III and IV at 12hrs post coinfection. The similar expression of 137

    BZLF-1 was determined in Infection-II and III at 24 hrs post-infection as compared to 138

    infection-IV. In 48hrs of post coinfection, there was a slightly higher expression of BZLF-1 139

    in infection-II concerning infection-III and IV Fig2A (III). Meanwhile, when comparing the 140

    infection-II, III, and IV with uninfected samples we observed that there was multiple fold 141

    higher expression of EBV associated latent and lytic genes, which confirmed the persistence 142

    of infection. 143

    Additionally, we investigated the expression of H. pylori-associated genes' transcripts such as 144

    16s-rRNA, pathogenic gene CagA, and BabA. Our finding showed that in the case of 145

    coinfection the expression of 16s-rRNA transcripts is higher as compared to individual 146

    infection of H. pylori (infection-I). The expression of 16s-rRNA transcripts is higher (~2000 147

    folds) in infection-III while it has downregulated in infection-IV at 12hrs post-infection. 148

    Moreover, the expression of this gene is about double in 24 and 48hrs as compared to 12hrs 149

    post-infection in infection-III and IV. In addition to this, the expression of 16s-rRNA 150

    transcripts increases successively from 12 (~2000 folds) to 48hrs (5000 folds) post-infection 151

    Fig2B (I). 152

    Further, we investigated the expression of well-known pathogenic genes such as CagA and 153

    BabA. Following the qRT-PCR results, we determined that the expression of CagA 154

    transcripts is significantly higher at 12 (p=0.0026 and 0.0020), 24 (p=0.0027, 0.0024), and 48 155

    (p=0.0023 and 0.0004) hrs time point in infection-III and IV as compared to infection-I and II 156

    Fig2B (II). Surprisingly, the expression of BabA is higher (~100-200 folds) at 12, 24, and 157

    48hrs time point in infection-I while it significantly decreases in infection-II, III, and IV in all 158

    the time points such as 12, 24, and 48 hrs post-infection Fig 2B (III). 159

    EBV and H. pylori regulate the expression of gankyrin in gastric epithelial cells 160

    In addition to pathogens' associated factors, we also determined the host factor that is 161

    regulated by the coinfection of EBV and H. pylori. Elevated expression of ankyrin was 162

    shown to be associated with several cancers. We investigated if this was also true for 163

    pathogen mediated gastric cancer. As expected, the infection of H. pylori and EBV enhanced 164

    the expression of host-associated oncogenic gene gankyrin at transcripts and protein levels. 165

    The expression of gankyrin is slightly higher at 12 and 24 hrs in infection-III and IV as 166

    compared to infection-I and II meanwhile, at 48 hrs post-infection there is significantly lower 167

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 5

    (p=0.0323, 0.0234, 0.0410 and 0.0415) expression of gankyrin in infection-III and IV 168

    whereas we have observed higher expression in infection-I and II Fig3A (I). Additionally, we 169

    have also observed a similar pattern of expression of gankyrin in western blot at protein level 170

    except for infection-III & IV where we examined the less expression of gankyrin protein as 171

    compared to infection-II after 12hrs of post-infection Fig3A (II) and (III). 172

    Coinfection of EBV and H. pylori promotes the oncogenic properties of epithelial cells 173

    by deregulating the expression of cell-cycle regulator and the tumor suppressor genes 174

    qRT-PCR results showed that CCND-1 gene expression is significantly higher in infection-III 175

    and IV at time point 12 (p=0.0231 and 0.0020) and 24 (p=0.0246 and 0.0349) hrs as 176

    compared to individual infected samples such as infection-I and II Fig4A (I&II). 177

    Surprisingly, we witnessed the significantly lower expression of CCND-1 at time point 48hrs 178

    in the infection-II, III, and IV in comparison to infection-I (p = 0.0021) Fig4A (III). In the 179

    case of DAPK3, after 12hrs post-infection the expression of this genes' transcripts was higher 180

    in infection-I and III as compared to infection-II and IV Fig4B (I) while in 24hrs post 181

    infected condition we observed the higher DAPK3 expression in infection-III and IV as 182

    compared to infection-I and II Fig4B (II) whereas adverse expression pattern was observed at 183

    48hrs post-infection. Further, we have observed lower expression of DAPK3 in all infected 184

    models in contrast to uninfected control Fig4B (III) 185

    The expression of another gene PCNA was higher in 12, 24, and 48hrs post infected samples 186

    concerning uninfected control, while a lower expression of this gene was examined at 48hrs 187

    in comparison to 12 and 24hrs post infected conditions in infection-III and IV Fig4C (I, II & 188

    III). The expression of another important gene Akt is comparatively higher in all infected 189

    models at 12 and 24hrs post-infection for 48hrs post infected samples Fig4D (I, II & III). 190

    Further, in the case of 24hrs, we witnessed the significantly higher expression of these genes' 191

    transcripts in infection-IV in comparison of infection-I and II (p= 0.0023, 0.0002, 0.0021 and 192

    0.0008) Fig4D (II) 193

    Tumor suppressor genes are the defense system of the body that helps to control the abnormal 194

    growth and proliferation of cells through various mechanisms such as promotes the DNA 195

    repair system, apoptosis, or modulates cell signaling. Expression of one of the important 196

    tumor suppressor gene PTEN is significantly higher in infection-I and II in comparison to 197

    infection-III and IV at 12 and 24hrs time points Fig4E (I & II). The expression of this gene 198

    is downregulated in all infection models as compared to uninfected control at 48hrs of post-199

    infection Fig4E (III). 200

    Additionally, APC is another tumor suppressor gene which performs their function through 201

    the interaction of cell adhesion molecules E-cadherin and negatively regulates the expression 202

    of beta-catenin. We have evidence that the expression of APC is significantly downregulated 203

    in the case of 12hrs post-infection in all the four infection models (p= 0.0231, 0.0248, 0.0349, 204

    and 0.0423) Fig4F (I). Unexpectedly, the expression of APC is upregulated in 24hrs post-205

    incubation of these two pathogens in infection-I, II, III, and IV while higher expression of 206

    this gene was observed in infection-I and II in comparison of infection-III and IV Fig4F (II). 207

    Moreover, in 48hrs post infected samples, we determined the higher expression of APC in 208

    infection-II followed by infection-I and IV Fig4F (III). 209

    Furthermore, we noticed the expression of the transcripts of another important tumor 210

    suppressor gene p53 was higher in infection-I and II in comparison to infection-III and IV at 211

    12 and 24hrs post infected conditions Fig4G (I & II). While in 48hrs post-infection, the 212

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 6

    expression of this gene was higher in infection-II concerning infection-I, III, and IV Fig4G 213

    (III). 214

    Similarly, pRb, a tumor suppressor gene, was showing its higher expression in infection-I in 215

    contrast to infection-II, III & IV at 12 and 24 hrs post-infection Fig4H (I & II). Meanwhile, 216

    the opposite expression pattern was observed in 48hrs of the infection Fig4H (III). 217

    H. pylori and EBV linked with the abrupt expression of gastric cancer marker and cell 218

    migratory genes by interfering with the oncogenic assessment of epithelial cells 219

    The expression of gastric cancer marker gene Abl-1 is significantly higher in infection-III 220

    (p=0.0210) as compared to infection-I and II at 12hrs post-infection whereas we witnessed 221

    the downregulation in case of infection-IV as compared to infection-I and II Fig5A (I). 222

    Further, its expression is higher in all infected models in comparison to uninfected control 223

    after 24 hrs of post-co-infection Fig5A (II). Surprisingly, the expression of Abl-1 is 224

    downregulated in the case of infection-II, III, and IV as compared to infection-I Fig5A (III). 225

    Adversely, the higher expression of TFF-2 was observed in infection-I and II in comparison 226

    with uninfected control and infection-III and IV Fig5B (I). In the case of 24hrs post-227

    infection, we determined the significantly higher expression of TFF-2 in infection-I followed 228

    by the infection-III and IV Fig 5B (II). Besides these, in 48hrs post, infected samples the 229

    expression of TFF-2 increased successively from infection-I to IV while the expression of 230

    this gene is comparatively alleviated in respect to 12 and 24hrs of post-infection Fig5B (III). 231

    Expression of another gastric cancer marker gene CDX-2 is higher in infection-I and III in 232

    comparison to infection-II and III at 12hrs of post infected samples Fig5C (I). CDX-2 233

    expression was slightly higher in infection-I as compared to other infection models in 24hrs 234

    post infected samples Fig5C (II). In addition to the above results it was significantly 235

    decreased in infection-I, II, III, and IV in 48hrs post-infected samples Fig5C (III). 236

    The upregulation of cell migratory genes is a hallmark for cancer cell metastasis. We 237

    witnessed the expression of MMP3, another cell migratory gene that helps the cells to 238

    metastasize, was also significantly upregulated in infection-III and IV in comparison of 239

    infection-I and II in 12, and 24hrs post infected samples Fig5D (I & II). Additionally, after 240

    48hrs post-infection of EBV and H. pylori the expression of MMP3 is moderately higher in 241

    infection-I while in infection-II, III, and IV the expression of these gene transcripts relatively 242

    similar to uninfected control Fig5D (III). Although the results revealed that expression of 243

    MMP7 is upregulated in infection-III and IV in contrast to infection-I and II at 12 and 24hrs 244

    of post-infection Fig5E (I, II), while we observed the adverse expression in 48hrs of post-245

    infection Fig5E (III). 246

    EBV and H. pylori-associated pathogenic gene expression could cause GC by regulating 247

    DNA damage response genes and anti-apoptotic gene Bcl-2 248

    The effective function of DNA damage response genes in humans is to provide stability and 249

    maintain its genome integrity. Moreover, cancer-causing infectious agents such as EBV and 250

    H. pylori dysregulated the status of DNA damage response transcripts by changing the milieu 251

    of the surrounding gastric epithelial cells. Interestingly, we determined the expression of 252

    DNA damage response gene Apaf-1 expression is slightly higher in infection-I in 12hrs post 253

    infected samples. A quite similar expression was observed in infection-II, III, and IV in 254

    comparison to uninfected control Fig5F (I). Furthermore, the expression of this gene at 24hrs 255

    is similar to the 12 hrs post infected samples in all the studied models except infection II 256

    where we observed higher expression of Apaf-1 Fig5F (II). Meanwhile, the expression of 257

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 7

    Apaf-1 in 48hrs post infected samples successively increases from the infection-I to IV Fig5F 258

    (III). 259

    In addition to Apaf-1, the expression of Bax is significantly downregulated in 12hrs post 260

    infected samples in all the conditions except in infection-II Fig5G (I). While the expression 261

    of Bax is successively downregulated in all the four infections in 24 hrs post infected samples 262

    Fig5G (II). Furthermore, this gene showed higher expression in infection-I, II, III, and IV in 263

    comparison of uninfected control in 48hrs time point, whereas, in infection-III and IV the 264

    expression of Apaf-1 was about 1.5-fold higher than the infection-I, II Fig5G (III). 265

    DNA damage response genes perform their function through its anti-apoptotic properties 266

    hence, the overexpression of anti-apoptotic genes such as Bcl-2 helps in cell survival and 267

    proliferation. Pathogens such as EBV and H. pylori modulate the expression of this type of 268

    host protein and promote the severity of gastric cancer. Coinfection of EBV and H. pylori 269

    upregulates the expression of anti-apoptotic gene Bcl-2 in 12hrs post-infection in all the four 270

    infection models compared to uninfected control whereas, we determined significantly higher 271

    expression of this gene in infection-IV Fig5H (I). In addition to 12hrs, the similar expression 272

    of Bcl-2 is observed in 24hrs post infected samples except for the infection-III in which we 273

    have detected the higher expression of Bcl-2 Fig5H (II). While in 48hrs post infected 274

    samples the expression of Bcl-2 is still higher compared to uninfected control. Moreover, the 275

    expression of this gene successively decreases from the infection-I to IV Fig 5H (III). 276

    Coinfection of EBV and H. pylori promotes GC through the upregulation of oncogenic 277

    protein gankyrin in a time-dependent manner 278

    Coinfection of EBV and H. pylori enhances the expression of oncogenic protein gankyrin in a 279

    time-dependent manner. As in the above-mentioned results, we revealed that the EBV and H. 280

    pylori could cause gastric cancer through the upregulation of gankyrin transcripts. To further 281

    support our qRT-PCR results we performed the immunostaining of gankyrin after coinfection 282

    of EBV and H. pylori by following the above-mentioned infection models (infection-I, II, III, 283

    and IV). Surprisingly, we obtained the similar expression pattern of gankyrin in 284

    immunostaining as observed at the transcript level. While, the higher expression of protein 285

    gankyrin is observed in all the four infectious models moreover, in infection-III and IV we 286

    evident that there is a significantly higher expression of gankyrin in 12hrs post-infected 287

    samples Fig6A (I&II). Interestingly, in 24hrs post infected samples the expression of 288

    gankyrin remains higher in infection-III and IV in comparison of infection-I and II Fig6B 289

    (I&II). Furthermore, we determined the significantly downregulated expression of gankyrin 290

    at 48hrs post infected samples in all four studied models in comparison to 12 and 24hrs post 291

    infected samples Fig6C (I&II). 292

    Endogenous and exogenous expression of gankyrin promotes the cell proliferation by 293

    dysregulating the cell migration, cell-cycle regulator, protooncogene, gastric cancer 294

    marker, and tumor suppressor gene in AGS cells 295

    Protein blot image showed the exogenous gradient expression of gankyrin in AGS cells after 296

    48hrs of post-transfection of the plasmid containing gankyrin gene Fig7 (A) and B. The 297

    result revealed a concentration-dependent increase in gankyrin expression. Moreover, to 298

    support our hypothesis that EBV and H. pylori cause gastric cancer via the upregulation of 299

    gankyrin we exogenously overexpressed gankyrin in AGS cells and performed cell 300

    proliferation assay. Surprisingly, results showed that the exogenous expression of gankyrin 301

    promotes cell proliferation Fig7 (C). Furthermore, to validate our previous results the 302

    coinfection of EBV and H. pylori upregulated the expression of oncogenic protein gankyrin 303

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 8

    which in turn changes the cell microenvironment and promotes cell proliferation we 304

    performed the cell proliferation assay by following all the four studied infection models. 305

    Interestingly, we observed that the rate of cell proliferation is comparatively significantly 306

    higher than the uninfected mock. Besides this in 12, 24, 36, and 48hrs post infected samples 307

    the number of viable cells is higher in infection-II, III, and IV in comparison to infection-I 308

    and mock Fig7 (D) 309

    Interestingly, exogenous overexpression of gankyrin results showed that the oncogene 310

    gankyrin play their role via the interaction of various host cellular physiological pathway as it 311

    acts differentially on a various cellular mechanism such as cell migration, cell-cycle 312

    regulator, proto-oncogenes, gastric cancer marker, and tumor suppressor genes. Exogenous 313

    overexpression of gankyrin significantly upregulated the cell migratory gene MMP7 and 314

    MMP3 Fig7 (E) and the cell cycle regulator transcripts of Akt and CCND-1 Fig7 (F). Fig7 315

    (G) Showed the ectopic expression of gankyrin which enhances the expression of Hurp. 316

    Interestingly, the expression of tumor suppressor genes PTEN, APC, and pRB significantly 317

    downregulated in exogenously overexpressed samples Fig7 (H). Furthermore, the ectopic 318

    overexpression of gankyrin increases the expression of gastric cancer marker gene CCL8, 319

    TFF-2, and CDX-2 while it downregulated the expression gastrin Fig7 (I). 320

    Coinfection of EBV and H. pylori promotes the cell migration linked with the gankyrin 321

    We further investigated whether the cellular physiological changes elicited by EBV and H. 322

    pylori could contribute to cell migration using a wound-healing assay. This assay 323

    demonstrated that EBV and H. pylori stimulated motility in the AGS cells. As expected, we 324

    observed the higher cell migration in infection-III and IV (p = 0.0025, 0.0023, 0.0022, and 325

    0.0027) Fig8A (I) in all the selected time points. Furthermore, we counted the migratory cell 326

    and represented in graph Fig8A (II) 327

    Coinfection and ectopic overexpression of gankyrin promotes the oncogenic properties 328

    of AGS cells 329

    We performed the clonogenic assay to select the EBV positive cells for the understanding of 330

    oncogenic transformation efficiency of coinfected samples in comparison to individual 331

    infection. The pore size of the transwell is smaller than the H. pylori only allowed the H. 332

    pylori secretory molecules to reach the AGS cells, not the H. pylori. The experimental 333

    models are depicted in Fig9 (A). The lower number of colonies was observed in uninfected 334

    and H. pylori-infected AGS cells. Besides these the density of colonies is more in EBV 335

    infected AGS cells followed by the significantly higher number of colonies were determined 336

    in coinfected samples (B). Further, we quantified these colony numbers into the density and 337

    observed that the density of colonies was higher in EBV and H. pylori coinfected samples 338

    (C). As we speculated, we observed that the secretory molecules which are secreted by H. 339

    pylori may be linked with the increased EBV virion in AGS cells. To further validate our 340

    finding the coinfection of EBV and H. pylori promote the oncogenic properties of AGS cells 341

    through the upregulation of gankyrin. We exogenously overexpressed the gankyrin in AGS 342

    cells and determined that the ectopic expression of gankyrin enhanced the clonogenic 343

    properties of AGS cells (D). We quantified the densities of colonies and represented them in 344

    graph (E). 345

    Discussion 346

    Chronic or acute infection of pathogens that destroy the immune system is a severe threat to 347

    human health and can cause diseases like Alzheimer's, meningitis, cancer, etc (19-21). 348

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 9

    Infectious agents not only induced the carcinogenesis but it is also involved in the promotion 349

    of its aggressiveness (22, 23). About one-fifth of total human oncogenesis is associated with 350

    infectious agents. Pathogens modulate the cellular milieu by dysregulation the gene 351

    expression, fit for the potential carcinogenesis (24, 25). Studies suggested that H. pylori and 352

    EBV promote oncogenesis by reframing the expression of cellular genes at the level of 353

    transcription and translation in infected cells (2). Reprogramming of expression of transcript 354

    and protein could promote the intrusiveness of these lesions. In this study, AGS cell lines 355

    were used which mimic the primary gastric epithelial cells except for adherent’s junction. It 356

    is well established that H. pylori and EBV infect the AGS cells and NCI-N87 in equal 357

    efficiency (2, 25). Thus, all these above reasons elicited us to investigate the synergistic 358

    effect of H. pylori exposed AGS cells for the successful infection of EBV and further how the 359

    microenvironment of EBV infected AGS cells favour the fortunate infection of H. pylori. 360

    Various studies reported that H. pylori exposed cellular environmental conditions are suitable 361

    for the enhanced EBV infection and vice-versa (2, 25). Moreover, we also observed the 362

    enhanced EBV-GFP fluorescence in fluorolog analysis at various time intervals in all the 363

    coinfection models except in infection-I (data not shown). In a study by Pandey et. al. and 364

    Sonkar et. al, reported that these pathogens dysregulated the cellular genes’ transcript 365

    expression which led to the aggressive GC (2). Moreover, till yet it is an enigma whether the 366

    prior exposures of H. pylori create a suitable niche for the EBV infection or anterior windage 367

    of EBV creates a niche that favors the aggressive growth of H. pylori and further gastric 368

    carcinoma. Interestingly, we got similar results as being speculated, coinfection of EBV and 369

    H. pylori enhanced the pathogenic gene expression such as EBNA-1, EBNA3C, and BZLF-1 370

    in various time intervals. Various studies reported that EBNA-1 and EBNA-3C enhanced the 371

    G1/S phase transition, cell invasiveness, and metastasis through the upregulation of cell 372

    signaling genes such as slug, snail, vimentin, and E-cadherin (23, 26-29). In addition to these 373

    functions, it is also known to regulate tumor suppressor genes like p53, pRb which play an 374

    important role in oncogenesis and cancer progression. 375

    In this study, we determined the expression of EBV associated latent genes (EBNA-1 and 376

    EBNA3C) can significantly promote the replication and spread of the virus in coinfected 377

    conditions up to 48hrs then the individual infection. The gene expression pattern in H. pylori 378

    and EBV coinfections revealed that H. pylori contributed to the increased replication and 379

    transcription of EBV pathogenic genes and virion production. Surprisingly, we observed that 380

    another latent gene EBNA3C expression is significantly correlated with the expression of 381

    oncogenic protein gankyrin at 12, 24, and 48hrs post infected samples. To support this 382

    finding, we performed the western blot analysis in which we observed the same expression 383

    pattern of gankyrin as we observed in qRT-PCR. These results revealed that the EBV-384

    associated EBNA3C may be directly linked with the expression of host oncogenic protein 385

    gankyrin as the expression of EBNA3C and gankyrin was alleviated in the only presence of 386

    EBV. The current study suggested that EBNA3C could directly regulate the host oncogenic 387

    protein gankyrin expression. These results supported that EBNA3C may cause gastric cancer 388

    through the regulation of host oncogenic protein gankyrin. It is an established oncoprotein 389

    which is found to be overexpressed in various cancers and involved in various biological 390

    processes, encompassing transformations of normal cells into cancerous one (15). Abnormal 391

    expression of gankyrin observed in human liver cancer, pancreatic cancer, oesophageal 392

    cancer, cervical, lung cancer, breast cancer, and glioma (16). Although there is no report 393

    available till which suggests that infectious agents caused the GC through the upregulation of 394

    oncogene gankyrin? 395

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 10

    This is the first study demonstrating that the exposure of AGS cells with EBV before H. 396

    pylori infection and vice-versa increased the expression of gankyrin which led to aggressive 397

    gastric carcinogenesis. The expression pattern of bacterial signature gene 16s-rRNA indicated 398

    the establishment of infection in AGS cells. H. pylori-associated secretory antigen CagA 399

    encoded by the CagA-pathogenicity island (CagA-PAI) present in about 89% of H. pylori 400

    strains caused morphological dysplastic alteration in epithelial cells (30), (31). Dysplastic 401

    changes are important and contribute to the initiation of carcinogenesis. Furthermore, H. 402

    pylori encoding CagA is associated with hustle inflammation, peptic ulcers, gastritis, and GC 403

    (24). 404

    In the present study, we determined that the human gastric epithelial cells have enhanced 405

    susceptibility to EBV infection in the presence of CagA positive H. pylori strain (I10). 406

    Pandey et al. showed that a CagA mutant strain of H. pylori has a significant reduction in 407

    viral titer production (2). This study was suggested that instead of CagA mutant, wild type H. 408

    pylori contributed significantly to viral replication and subsequently cancer progression and 409

    aggressiveness. H. pylori loaded with type-IV bacterial secretion system that helps in the 410

    injection of its associated secretory protein into the host-derived cells. Alternative mode of 411

    uptake of CagA through integrins mediated rearrangements of actin filaments and further 412

    endocytosis. Endocytosis of CagA further changed the intracellular milieu and led to the 413

    morphological alteration in the gastric epithelial cells. Here, our results supported the 414

    hypothesis that the type-IV secretory mechanism was used by this bacterium, as the direct 415

    incubation of wild type H. pylori showed enhanced infectivity. The expression of signature 416

    sequence 16s-rRNA, CagA were higher in coinfected samples which concluded that the virus 417

    could also help synergistically to increase the expression of H. pylori associated factors. 418

    Expression of 16s-rRNA inferences that enhanced replication and multiplication of H. pylori 419

    genome in presence of EBV. BabA transcripts are known to be highly expressed in peptic 420

    ulcer and gastric cancer rather than asymptomatic colonization. Similar observations were 421

    made with the BabA transcript in infection-I which showed that the expression of BabA is 422

    important for enhanced infectivity. Further, the higher BabA expression early point suggested 423

    the increased infectivity of H. pylori as it helps in the attachment of H. pylori with host cells. 424

    In addition to the above results, the recruitment of gankyrin by the coinfection dysregulated 425

    the downstream genes which were known to be directly influenced by upregulation of this 426

    protein. These genes are studied under six categories such as cell cycle regulator genes, GC 427

    marker, cell migratory, tumor suppressor, DNA damage response, and antiapoptotic gene. 428

    Moreover, we investigated that EBNA-3C and gankyrin can further change the status of 429

    various cellular proteins which have played a significant role in gastric carcinogenesis. 430

    Although, experiments need to be conducted to validate the one-to-one relation of EBNA3C 431

    and gankyrin. Results of the present study suggested that the differential expression of cell 432

    cycle regulatory genes such as CCND-1, DAPK3, PCNA, and Akt in all the infectious 433

    models at various time points could be linked with the increased oncogenesis. Gankyrin 434

    upregulated the CCND-1 and PCNA in pancreatic and gastric cancer respectively (32). 435

    Besides these genes, the expression of gankyrin upregulates the Akt at transcript and protein 436

    level in hepatocellular, liposarcoma, and gastric cancer (33–35). The upregulation of Akt 437

    protein through the action of gankyrin enhanced cell proliferation, metastasis, stem cell self-438

    renewal capacity, autophagy, and chemoresistance (18, 36). The upregulation of death-439

    associated protein kinase domain-3 (DAPK-3) is linked with enhanced apoptosis. While 440

    coinfection of these two oncogenic pathogens downregulated this, pro-apoptotic which 441

    supported the accumulation of AGS gastric epithelial cells over the period. Results showed a 442

    significantly lower expression of gankyrin in 12 and 24hrs post-infection in comparison of 443

    48hrs. Furthermore, this finding supported that H. pylori and EBV infection mediated higher 444

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 11

    expression of gankyrin led to aggressive GC through the dysregulation of cell-cycle 445

    regulatory genes. 446

    EBV and H. pylori can hijack one of the GC markers non-receptor tyrosine kinase (Abl-1) 447

    signaling to remodel the host cytoskeleton for multiple purposes such as late phase 448

    autophagy, cell motility, and cell adhesion. Trefoil factor-2 (TFF-2) is another gene that is 449

    known to be overexpressed in GC. It is a structural component of gastric mucosa and inhibits 450

    gut acid secretion and its motility. Inhibition of acid secretion provides the platform for the 451

    aggressive growth of H. pylori which can further enhance the EBV reactivation and 452

    replication (37, 38). Homeobox protein (CDX-2) preferentially binds to methylated DNA and 453

    is involved in multiple cellular processes such as transcriptional regulation of genes of 454

    epithelial cells. In the present study, profiles of GC marker genes suggested that H. pylori 455

    changed the status of GC related marker genes for their growth and development of 456

    oncogenesis. It was not only involved in the dysregulation of host oncogenic factors but also 457

    changed the expression of genes that provided a microenvironment for its growth and 458

    replication. 459

    Matrix metalloproteinase-3 and 7 (MMP-3 & MMP-7) is an enzyme that increases the 460

    expression of pro-collagenase and degrades the collagen layer, fibronectin, and laminin. The 461

    overexpression of this protein in infection models indicates the probable role of EBV and H. 462

    pylori in gankyrin mediated aggressiveness of GC through the recruitment of such kinds of 463

    cell migratory genes. 464

    Downregulated form of the phosphatase and tensin homolog (PTEN) is associated with 465

    several types of cancers and functions as a negative regulator of the PI3-kinase/AKT pathway 466

    by downregulating PIP3 (phosphatidylinositol 3,4,5-triphosphate) levels. The protein 467

    gankyrin downregulated the tumor suppressor protein-16 (p-16), protein-53 (p-53), and 468

    protein retinoblastoma (pRB) through the direct interaction with its various domains (17). 469

    The loss of activity of these tumor suppressor genes is the key marker for carcinogenesis. 470

    This demonstrates that the association of infection of EBV and H. pylori in gankyrin 471

    upregulation, cellular genome, and dysregulation of cellular signaling was important for 472

    oncogenesis. 473

    Apaf-1 and Bax are mitochondrial enzymes that lead to the release of cytochrome c and 474

    CASP3 and efficiently initiate the process of apoptosis. Apoptotic protease activating-factor-475

    1 (Apaf-1) and apoptosis regulator (Bax) profiles after coinfection suggested that the 476

    coinfection of EBV and H. pylori downregulated the proapoptotic genes via upregulating the 477

    oncoprotein gankyrin. Furthermore, EBV and H. pylori infection mediated upregulated 478

    gankyrin downregulates the anti-apoptotic B-cell lymphoma-2 (BCL-2). 479

    The current study demonstrated that EBV and H. pylori-associated factors specifically 480

    enhanced the expression of gankyrin. Upregulated gankyrin mediated dysregulation of 481

    expression of the cell-cycle regulator, GC marker, cell migration, DNA response, and 482

    antiapoptotic genes in infected gastric epithelial cells. EBV and H. pylori create intracellular 483

    and extracellular milieu which potentially favor gastric epithelial cell proliferation and 484

    carcinogenesis. It provided the H. pylori-EBV tie, which forms a suitable tumor 485

    microenvironment for gastric epithelial cell proliferation and carcinogenesis. Further 486

    gankyrin might be playing a central role in this transformation process by dysregulation of 487

    multiple associated genes Fig 10. 488

    489

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 12

    Materials and methods 490

    Cell culture: 491

    The gastric cell lines AGS were obtained from the National Center for Cell Science (NCCS) 492

    Pune India. AGS cell line which is an EBV negative cells, cultured in Dulbecco’s modified 493

    Eagle’s medium (DMEM; Himedia, Mumbai, India) containing 10% fetal bovine serum 494

    (FBS; BIOWEST, South America origin) and 100 U/mL penicillin/streptomycin (Himedia, 495

    Mumbai India) under conditions of 5% CO2 and humidified air at 37°C (Eppendorf India). 496

    Virus (EBV) culture and isolation: 497

    HEK293T cells containing GFP-EBV (obtained as a gift from the Erle S. Robertson’s 498

    laboratory, University of Pennsylvania, USA) were cultured and used for EBV amplification. 499

    Cultured the virus and purified as described previously (2, 39, 25) 500

    Bacterial (H. pylori) culture: 501

    H. pylori strain (I10) was a kind gift by Dr. Asish Kumar Mukhopadhyay (National Institute 502

    of Cholera and Enteric Diseases, ICMR, Kolkata, India) H. pylori was grown according to the 503

    conditions described earlier (25). 504

    Coinfection of EBV and H. pylori with AGS cells: 505

    For infection, we took 0.25 million AGS cells and cultured in a 6-well plate. Observed the 506

    cells up to the 40-50% confluencies and then incubated the MOI-100 of H. pylori and 25μl of 507

    EBV. We optimized the EBV dose by incubating the increasing dose of EBV such 25, 50, 508

    100, 150, and 200 μl with AGS cells and observed 25 μl of EBV culture was sufficient for a 509

    successful infection. For this study, we have developed five independent approaches for the 510

    construction of coinfection models by using H. pylori and EBV. In the first approach, 511

    uninfected AGS cells were taken as the negative control (Fig.1A control). In our second and 512

    third approaches, we have incubated the H. pylori and EBV with AGS cells (Fig.1B 513

    infection-I and C Infection-II) respectively. Moreover, in the fourth approach, we have 514

    incubated the H. pylori with AGS cells for six hours thereafter given the infection of EBV 515

    and incubated for 12, 24, and 48 hours (Fig.1D infection-III). In the fifth approach, the 516

    infection of EBV is given first for six hours followed by the infection of H. pylori (Fig.1E 517

    Infection-IV). 518

    RNA isolation and qRT-PCR: 519

    Fixed (MOI-100) of H. pylori (I10) and EBV (25μl) were incubated with AGS cells under 520

    specific conditions (5% CO2, 37 °C) for 12, 24, and 48 hrs. RNA isolated and qRT-PCR was 521

    executed as described earlier (25). The pathogens and host genes were analyzed. 522

    Western Blot: 523

    Western blot experiments were carried out as described earlier (40). The lysates were 524

    analyzed by Western blots using an anti-gankyrin polyclonal primary antibody (cell 525

    signaling) and HRP-tagged secondary antibody. Blots were observed under the gel doc 526

    system (ChemiDoc™ XRS+ System with Image Lab™ Software #1708265) 527

    Immunofluorescence Assay 528

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 13

    An immunofluorescence assay was performed as described earlier (40) by using an anti-529

    gankyrin primary antibody and specific signals were detected with secondary antibodies 530

    conjugated with Alexa Fluor 488 (cell signaling). The cell's nucleus was counterstained with 531

    4′, 6′-diamidino-2-phenylindole (DAPI). The results were analyzed with confocal microscopy 532

    (Olympus IX83) at 60X and zoom two times in triplicates. 533

    Cell proliferation assay 534

    An assay of cell proliferation was accomplished through trypan blue exclusion and crystal 535

    violet staining methods as described earlier (2, 40). 536

    537

    Colony formation assay 538

    The AGS cells were infected with EBV and H. pylori and EBV positive cells selected with 539

    puromycin (2ug/ml). We selected the ectopically expressed gankyrin positive AGS cells 540

    through geneticin (G148). The image was taken by using ChemiDoc™ XRS+ System with 541

    Image Lab™ Software #1708265 (40). 542

    Statistical Analysis 543

    Data were statistically analyzed using a two-tailed Student's t-test. All the results were 544

    derived from triplicate experiments P-values by using the Graph Pad Prism version 6 of 545

  • 14

    H. pylori (Helicobacter pylori); EBV (Epstein Barr Virus); GC (Gastric cancer); EBNA1 567

    (Epstein Barr Nuclear antigen-1); EBNA3C (Epstein Barr Nuclear antigen-3C); EBNA2 568

    (Epstein Barr Nuclear antigen-2); BZLF-1 (BamHI Z fragment leftward open reading frame 569

    1); 16s rRNA (16s Ribosomal RNA); CagA (cytotoxin-associated gene A); BabA (Blood 570

    group antigen binding adhesin A); VacA (Vacuolating toxin A); pRb (protein 571

    retinoblastoma); P53 (Tumor suppressor protein 53); AGS (Adenogastric carcinoma cell); 572

    CCND-1 (Cyclin D1); DAPK3 (Death associated protein kinase domain-3); PCNA 573

    (Proliferating cell nuclear antigen); Akt (Protein kinase B); PTEN (Phosphatase and tensin 574

    homolog); APC (Adenomatous polyposis coli); Abl-1 (Tyrosine-protein kinase ABL1); TFF2 575

    (Trefoil factor-2); CDX-2 (Homeobox protein CDX-2); MMP-3 (Matrix metalloproteinase-576

    3); MMP-7 (Matrix metalloproteinase-7); Apaf-1 (Apoptotic protease activating-factor-1); 577

    Bax (Apoptosis regulator BAX); Bcl-2 (B-cell lymphoma-2); CCL8 (Chemokine ligand 8); 578

    CagA-PAI (CagA-pathogenicity island); PIP3 (phosphatidylinositol 3,4,5-triphosphate) 579

    580

    References 581

    1. Sitarz R, Skierucha M, Mielko J, Offerhaus GJA, Maciejewski R, Polkowski WP. 582

    2018. Gastric cancer: epidemiology, prevention, classification, and treatment. Cancer Manag 583

    Res 10:239–248. 584

    2. Pandey S, Jha HC, Shukla SK, Shirley MK, Robertson ES. 2018. Epigenetic 585

    Regulation of Tumor Suppressors by Enhances EBV-Induced Proliferation of Gastric 586

    Epithelial Cells. MBio 9:e00649-18. 587

    3. de Souza CRT, Almeida MCA, Khayat AS, da Silva EL, Soares PC, Chaves LC, 588

    Burbano RMR. 2018. Association between Epstein-Barr virus, human papillomavirus and 589

    gastric adenocarcinomas. World J Gastroenterol 24:4928–4938. 590

    4. Salih BA. 2009. Helicobacter pylori infection in developing countries: the burden for 591

    how long? Saudi J Gastroenterol 15:201–207. 592

    5. Brown LM. 2000. Helicobacter pylori: epidemiology and routes of transmission. 593

    Epidemiol Rev 22:283–297. 594

    6. Rizzato C, Torres J, Plummer M, Muñoz N, Franceschi S, Camorlinga-Ponce M, 595

    Fuentes-Pananá EM, Canzian F, Kato I. 2012. Variations in Helicobacter pylori cytotoxin-596

    associated genes and their influence in progression to gastric cancer: implications for 597

    prevention. PLoS One 7:e29605. 598

    7. Ohnishi N, Yuasa H, Tanaka S, Sawa H, Miura M, Matsui A, Higashi H, Musashi M, 599

    Iwabuchi K, Suzuki M, Yamada G, Azuma T, Hatakeyama M. 2008. Transgenic expression 600

    of Helicobacter pylori CagA induces gastrointestinal and hematopoietic neoplasms in mouse. 601

    Proc Natl Acad Sci U S A 105:1003–1008. 602

    8. Del Moral-Hernández O, Castañón-Sánchez CA, Reyes-Navarrete S, Martínez-603

    Carrillo DN, Betancourt-Linares R, Jiménez-Wences H, de la Peña S, Román-Román A, 604

    Hernández-Sotelo D, Fernández-Tilapa G. 2019. Multiple infections by EBV, HCMV and 605

    Helicobacter pylori are highly frequent in patients with chronic gastritis and gastric cancer 606

    from Southwest Mexico: An observational study. Medicine 98:e14124. 607

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 15

    9. Ayee R, Ofori MEO, Wright E, Quaye O. 2020. Epstein Barr Virus Associated 608

    Lymphomas and Epithelia Cancers in Humans. J Cancer 11:1737–1750. 609

    10. Mui UN, Haley CT, Tyring SK. 2017. Viral Oncology: Molecular Biology and 610

    Pathogenesis. J Clin Med Res 6:111. 611

    11. Knight JS, Sharma N, Kalman DE, Robertson ES. 2004. A cyclin-binding motif 612

    within the amino-terminal homology domain of EBNA3C binds cyclin A and modulates 613

    cyclin A-dependent kinase activity in Epstein-Barr virus-infected cells. J Virol 78:12857–614

    12867. 615

    12. Nishikawa J, Imai S, Oda T, Kojima T, Okita K, Takada K. 1999. Epstein-Barr virus 616

    promotes epithelial cell growth in the absence of EBNA2 and LMP1 expression. J Virol 617

    73:1286–1292. 618

    13. Takada K. 2000. Epstein-Barr virus and gastric carcinoma. Mol Pathol 53:255–261. 619

    14. Zamani P, Matbou Riahi M, Momtazi-Borojeni AA, Jamialahmadi K. 2018. 620

    Gankyrin: a novel promising therapeutic target for hepatocellular carcinoma. Artif Cells 621

    Nanomed Biotechnol 46:1301–1313. 622

    15. Li J, Guo Y. 2010. Gankyrin Oncoprotein: Structure, Function, and Involvement in 623

    Cancer. Curr Chem Biol 4:13–19. 624

    16. Li H, Zhang J, Zhen C, Yang B, Feng L. 2018. Gankyrin as a potential target for 625

    tumor therapy: evidence and perspectives. Am J Transl Res 10:1949–1960. 626

    17. Dawson S, Higashitsuji H, Wilkinson AJ, Fujita J, Mayer RJ. 2006. Gankyrin: a new 627

    oncoprotein and regulator of pRb and p53. Trends Cell Biol 16:229–233. 628

    18. Zeng Y-C, Sun D, Li W-H, Zhao J, Xin Y. 2017. Gankyrin promotes the proliferation 629

    of gastric cancer and is associated with chemosensitivity. Tumour Biol 630

    39:1010428317704820. 631

    19. Nicholson LB. 2016. The immune system. Essays Biochem 60:275–301. 632

    20. Baldwin KJ, Zunt JR. 2014. Evaluation and treatment of chronic meningitis. 633

    Neurohospitalist 4:185–195. 634

    21. Zhang J, Wei J, Wang Z, Feng Y, Wei Z, Hou X, Xu J, He Y, Yang D. 2020. 635

    Transcriptome hallmarks in Helicobacter pylori infection influence gastric cancer and MALT 636

    lymphoma. Epigenomics 12:661-671. 637

    22. Naseem M, Barzi A, Brezden-Masley C, Puccini A, Berger MD, Tokunaga R, 638

    Battaglin F, Soni S, McSkane M, Zhang W, Lenz H-J. 2018. Outlooks on Epstein-Barr virus 639

    associated gastric cancer. Cancer Treat Rev 66:15–22. 640

    23. Jha HC, Lu J, Saha A, Cai Q, Banerjee S, Prasad MAJ, Robertson ES. 2013. 641

    EBNA3C-mediated regulation of aurora kinase B contributes to Epstein-Barr virus-induced 642

    B-cell proliferation through modulation of the activities of the retinoblastoma protein and 643

    apoptotic caspases. J Virol 87:12121–12138. 644

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 16

    24. Wroblewski LE, Peek RM Jr, Wilson KT. 2010. Helicobacter pylori and gastric 645

    cancer: factors that modulate disease risk. Clin Microbiol Rev 23:713–739. 646

    25. Sonkar C, Verma T, Chatterji D, Jain AK, Jha HC. 2020. Status of kinases in Epstein-647

    Barr virus and Helicobacter pylori Coinfection in gastric Cancer cells. BMC Cancer 20:925. 648

    26. Gaur N, Gandhi J, Robertson ES, Verma SC, Kaul R. 2015. Epstein–Barr virus latent 649

    antigens EBNA3C and EBNA1 modulate epithelial to mesenchymal transition of cancer cells 650

    associated with tumor metastasis. Tumor Biology 36:3051-60. 651

    27. Davenport MG, Pagano JS. 1999. Expression of EBNA-1 mRNA is regulated by cell 652

    cycle during Epstein-Barr virus type I latency. J Virol 73:3154–3161. 653

    28. Nayyar VK, Shire K, Frappier L. 2009. Mitotic chromosome interactions of Epstein-654

    Barr nuclear antigen 1 (EBNA1) and human EBNA1-binding protein 2 (EBP2). J Cell Sci 655

    122:4341–4350. 656

    29. Jha HC, Yang K, El-Naccache DW, Sun Z, Robertson ES. 2015. EBNA3C regulates 657

    p53 through induction of Aurora kinase B. Oncotarget 6:5788–5803. 658

    30. Slater E, Owen RJ, Williams M, Pounder RE. 1999. Conservation of the cag 659

    pathogenicity island of Helicobacter pylori: Associations with vacuolating cytotoxin allele 660

    and IS605 diversity. Gastroenterology 117:1308–1315. 661

    31. Yamaoka Y. 2010. Mechanisms of disease: Helicobacter pylori virulence factors. Nat 662

    Rev Gastroenterol Hepatol 7:629–641. 663

    32. Meng Y, He L, Guo X, Tang S, Zhao X, Du R, Jin J, Bi Q, Li H, Nie Y, Liu J, Fan D. 664

    2010. Gankyrin promotes the proliferation of human pancreatic cancer. Cancer Lett 297:9–665

    17. 666

    33. Fu X-Y, Wang H-Y, Tan L, Liu S-Q, Cao H-F, Wu M-C. 2002. Overexpression of 667

    p28/gankyrin in human hepatocellular carcinoma and its clinical significance. World J 668

    Gastroenterol 8:638–643. 669

    34. Man J-H, Liang B, Gu Y-X, Zhou T, Li A-L, Li T, Jin B-F, Bai B, Zhang H-Y, Zhang 670

    W-N, Li W-H, Gong W-L, Li H-Y, Zhang X-M. 2010. Gankyrin plays an essential role in 671

    Ras-induced tumorigenesis through regulation of the RhoA/ROCK pathway in mammalian 672

    cells. J Clin Invest 120:2829–2841. 673

    35. Hwang J-A, Yang H-M, Hong D-P, Joo S-Y, Choi Y-L, Park J-H, Lazar AJ, Pollock 674

    RE, Lev D, Kim SJ. 2014. Gankyrin is a predictive and oncogenic factor in well-675

    differentiated and dedifferentiated liposarcoma. Oncotarget 5:9065–9078. 676

    36. Jing H, Zhang G, Meng L, Meng Q, Mo H, Tai Y. 2014. Gradually elevated 677

    expression of Gankyrin during human hepatocarcinogenesis and its clinicopathological 678

    significance. Sci Rep 4:5503. 679

    37. El-Omar EM. 2006. Mechanisms of increased acid secretion after eradication of 680

    Helicobacter pylori infection. Gut 55:144-146. 681

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 17

    38. Minoura-Etoh J, Gotoh K, Sato R, Ogata M, Kaku N, Fujioka T, Nishizono A. 2006. 682

    Helicobacter pylori-associated oxidant monochloramine induces reactivation of Epstein–Barr 683

    virus (EBV) in gastric epithelial cells latently infected with EBV. Journal of Medical 684

    Microbiology 55:905-911. 685

    39. Halder S, Murakami M, Verma SC, Kumar P, Yi F, Robertson ES. 2009. Early events 686

    associated with infection of Epstein-Barr virus infection of primary B-cells. PLoS One 687

    4:e7214. 688

    40. Jha HC, Sun Z, Upadhyay SK, El-Naccache DW, Singh RK, Sahu SK, Robertson ES. 689

    2016. KSHV-Mediated Regulation of Par3 and SNAIL Contributes to B-Cell Proliferation. 690

    PLoS Pathog 12:e1005801. 691

    692

    693

    694

    Figures and figure legends 695

    696

    Fig.1 Coinfection models of H. pylori and EBV depicts the deep into the pathophysiology 697 of gastric cancer. Illustration of all the studied infectious models. AGS cells were cultured in 698

    6-well plates, and directly incubated the H. pylori and EBV with these cells (A) Uninfected 699

    AGS control cells (B) Infection-I portrayed the infection of H. pylori (I10) (C) Infection-700

    II AGS were infected with EBV only. (D) Infection-III AGS cells were infected 701

    sequentially, first exposing the cells with EBV for 6 hrs then these exposed cells incubated 702

    with H. pylori. (E) Infection-IV AGS cells were firstly exposed with the H. pylori then 703

    incubated with EBV. All the infected cells were further incubated for 12, 24 and 48 hrs. 704

    These infection models were used for the whole of this study except colony formation assay. 705

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 18

    706

    Fig.2 Interplay between H. pylori and EBV synergistically increases the transcript of its 707 associated pathogenic genes. (A) Transcript expression of EBV associated latent and lytic 708

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 19

    genes. (I) & (II) Quantification of transcript of latent gene EBNA-1 & EBNA-3C respectively 709

    (III) qRT-PCR quantification of lytic gene BZLF-1 (B) Transcript quantification of H. pylori 710

    associated signature 16S-rRNA (I) and its associated pathogenic gene cytotoxin associated 711

    gene A (CagA) (II) and blood group antigen binding adhesin A (BabA) (III) in all the 712

    infection model for 12, 24 and 48 hrs post infected samples. The experiment performed for 713

    two biological and two technical replicates and the results are shown as the mean SD of two 714

    independent experiments. 715

    716

    Fig.3 Coinfection of H. pylori and EBV upregulates the oncogenic protein gankyrin at 717 both transcripts and protein levels (A) Transcript expression of gankyrin in coinfection 718

    models (I), (II) & (III) depict the expression of gankyrin at 12, 24 and 48 hrs respectively. (B) 719

    (I), (II) & (III) western blot image of protein gankyrin for 12, 24 and 48 hrs post infected 720

    samples respectively. (C) (I), (II) & (III) Illustrate the quantification of blot image by Image 721

    J. software and representative graph presented in terms of fold changes for 12, 24 and 48 hrs 722

    post infected samples respectively. The significantly higher expression of gankyrin observed 723

    at 12 and 24 hrs post infected samples. While the expression of gankyrin is significantly 724

    downregulated in infection-II, III and IV in 48 hrs post infected samples. The experiment was 725

    performed for two biological and one technical replicate. 726

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 20

    727

    728

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 21

    Fig.4 Coinfection of H. pylori and EBV promotes gastric cancer through the 729 dysregulations of cell-cycle regulator and the expression of tumor suppressor gene (A) I, 730

    II & III represents the cyclin D1 (CCND-1) profiles in coinfection at 12, 24 and 48 hrs 731

    respectively (B) I, II & III depicts the death associated protein kinase-3 (DAPK3) expression 732

    pattern at 12, 24 and 48 hrs respectively (C) I, II & III demonstrate the transcript expression 733

    profiles of proliferating cell nuclear antigen (PCNA) 12, 24 and 48 hrs respectively (D) I, II 734

    & III depicts the expression profiles of AKR thymoma (Akt) coinfected samples at 12, 24 and 735

    48 hrs of post infection of EBV and H. pylori. (E) I, II & III illustrate the downregulation of 736

    phosphatase and tensin homolog (PTEN) in 24 and 48 hrs post infection, while it is slightly 737

    upregulated in 12 hrs post infection. (F) I, II & III demonstrate the expression profiles of 738

    adenomatous polyposis coli (APC) in EBV and H. pylori coinfected samples for 12, 24 and 739

    48 hrs time points. (G) & (H) I, II & III represents the expression of p53 and pRB 740

    respectively in 12, 24 and 48 hrs post infected samples. The experiment has performed for 741

    two biological and two technical replicates and the results are shown as the mean SD of two 742

    independent experiments. 743

    744

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 22

    745

    Fig.5 Coinfection of H. pylori and EBV changes oncogenic assessment of gastric 746

    epithelial cells by dysregulating gastric cancer marker, cell migratory and antiapoptotic 747

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 23

    genes. (A) I, II & III depicts the status of tyrosine protein kinase Abl-1 gene expression in 748

    EBV and H. pylori coinfection. (B) I, II & III demonstrate the expression profiles of trefoil 749

    factor-2 (TFF-2) at 12, 24 and hrs time points. (C) I, II & III illustrate the transcription factor 750

    CDX-2 in coinfected samples for 12, 24 and 48 hrs of post infection. (D) I, II & III depicts 751

    the expression pattern of matrix metalloprotease-3 (MMP-3) in 12, 24 and 48 hrs post 752

    infected samples. (E) I, II & III represents the expression status of matrix metalloprotease-7 753

    (MMP-7) (F) I, II & III illustrate the downregulation of Apaf-1 (G) I, II & III represents the 754

    decrease in amount of Bax transcripts in coinfected samples at 12, 24 and 48 hrs of post 755

    infected samples. (H) I, II & III represents the expression pattern of BCL-2 in which at 12 756

    and 24 hrs post infection the expression of antiapoptotic gene is elevated as the expression of 757

    gankyrin is also several folds upregulated in 12 and 24 hrs post infected samples in above 758

    mentioned results. While the expression of BCL-2 follows the same pattern at 48hrs post 759

    infected as the expression of gankyrin was observed. The experiment has performed for two 760

    biological and two technical replicates and the results are shown as the mean SD of two 761

    independent experiments. 762

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 24

    763

    764

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 25

    Fig.6 Coinfection of EBV and H. pylori causes gastric cancer through the upregulation 765

    of oncogenic protein gankyrin. 766

    Immunofluorescence results of oncogenic gene gankyrin validate our previous transcripts and 767

    western blot results. Graphical representation of quantified immunoblotting image through 768

    the Image J. software. After coinfection of EBV and H. pylori we observed elevated protein 769

    expression of gankyrin in all the time points for all studied infection except 48 hrs post 770

    infection in which observed downregulation of gankyrin in infection-II, III and IV. (A) I, (B) 771

    I & (C) I Representative immunoblotting image of gankyrin represents the protein expression 772

    of gankyrin after 12, 24 and 48 hrs of post infection respectively. Graphical representation of 773

    quantified immunoblotting image through the Image J. software. First panel showed an 774

    uninfected mock. Second and third panel illustrate the gankyrin protein after I10 and EBV 775

    infection. While the fourth and fifth panel shows the intensity of gankyrin expression after 776

    EBV-I10 and I10-EBV post infection. (A) II Approximately 600-700-fold increased 777

    expression of gankyrin observed in EBV-I10 and I10-EBV coinfected samples as compared 778

    to individual infection of I10 and EBV after 12 hrs post infection. B (II) The expression 779

    pattern of gankyrin in 24 hrs post infected samples is about 100 to 200-fold higher in EBV-780

    I10 and I10-EBV coinfection. C (III) The expression of oncogenic protein gankyrin after 48 781

    hrs of post infection is about 3-32 higher in infection III and IV in comparison of infection-I. 782

    The expression of gankyrin in infection-II is about equal to infection III while the 25-30-fold 783

    lower expression of gankyrin was observed in infection IV. The experiment has performed 784

    for two biological and three technical replicates and the results are shown as the mean SD of 785

    two independent experiments. 786

    787

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 26

    788

    Fig.7 Coinfection mediated upregulation of gankyrin and ectopic expression of gankyrin 789

    promotes the rate of cell proliferation by interfering with the expression of various 790 cellular genes. (A) Western blot image represents the ectopic expression of oncogenic 791

    protein gankyrin. (B) Graphical representation of increased concentration gradient of 792

    gankyrin quantified by the image J software of western blot image. (C) Shows the ectopic 793

    expression of gankyrin enhances the cell proliferation in trypan blue cell exclusion method of 794

    cell counting (D) Moreover, coinfection of EBV and H. Pylori also enhances the rate of cell 795

    proliferation in coinfected samples and may be linked with the increased expression of 796

    gankyrin. (E) Represents the ectopic expression of gankyrin significantly enhanced the 797

    expression of cell migratory gene MMP-7 and MMP-3. (F) & (G) Representative graph 798

    shows the elevated expression of cell-cycle regulatory gene Akt and CCND-1 and proto-799

    oncogenes hepatoma upregulated protein (Hurp) respectively. Moreover, overexpression of 800

    gankyrin significantly alleviates the expression of tumor suppressor gene PTEN, APC and 801

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 27

    pRB shown in (H). While the expression of gastric cancer marker gene gastrin expression is 802

    slightly lower followed by the upregulation of C-C motif chemokine ligand-8 (CCL-8), TFF-803

    2 and CDX-2 (I). The experiment has performed for two biological and two technical 804

    replicates and the results are shown as the mean SD of two independent experiments. 805

    806

    807

    Fig.8 Coinfection of EBV and H. pylori enhanced the efficiency of cell migration of AGS 808 cells. (A) I Representative image of scratch wound healing assay. First panel shows the 809

    uninfected AGS cells. Second, third, fourth and fifth panel shows the infection-I, II, III and 810

    IV respectively for the 0, 6, 12, 24 and 48 hrs of post infection. (A) II Graphical 811

    representation of the number of cells migrated in each infection and uninfected mock. 812

    813

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 28

    814

    Fig.9 Coinfection and ectopic overexpression of gankyrin can be linked with the 815 elevated oncogenic properties of AGS cells. A (I) Uninfected AGS cells. (II) AGS cells 816

    with transwell and H. pylori infection. (III) AGS cell only with EBV infection. (IV) First 817

    infection of H. pylori for 6 hrs followed by second infection of EBV. (V) First infection of 818

    EBV for 6 hrs followed by second infection of H. pylori. Selection of EBV positive colonies 819

    for 14 days. (B) Representative image of foci formation after coinfection. (C) 820

    Quantification and graphical representation of density of foci through the Image J. software. 821

    (D) Further we have validated foci formation results through the ectopic expression of 822

    gankyrin in AGS cells and selected the gankyrin positive cells through the geneticin (G-418) 823

    for 14 days. Representative image of foci in exogenously overexpressed AGS cells. (E) 824

    Quantification and graphical representation of density of foci in exogenously overexpressed 825

    AGS cells through the Image J. software. The experiment has been performed three times 826

    and the results are shown as the mean SD of two independent experiments. 827

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 29

    828

    Figure 10: A model illustrating the association of gankyrin in EBV and H. pylori 829 mediated gastric cancer. This association drives the progression of EBV and H. pylori 830

    mediated gastric cell transformation. Oncogenic activity of gankyrin is accelerated in the 831

    presence of pathogen which targets the cell cycle regulators, cell migratory genes, gastric 832

    cancer marker, anti-apoptotic, DNA damage response and tumor suppressor genes thus 833

    caused the aggressive oncogenesis. 834

    835

    Supplementary Figure and figure legend 836

    837

    838

    Supplementary Fig.1 Gram staining of H. pylori (I10) showing the regular spiral shape of 839

    bacteria 840

    841

    .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4.0/

  • .CC-BY-NC-ND 4.0 International licenseperpetuity. It is made available under apreprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in

    The copyright holder for thisthis version posted November 20, 2020. ; https://doi.org/10.1101/2020.11.19.390807doi: bioRxiv preprint

    https://doi.org/10.1101/2020.11.19.390807http://creativecommons.org/licenses/by-nc-nd/4