Genetic Diversity of the cagA gene of Helicobacter pylori strains from Sudanese Patients with Different Gastroduodenal Diseases =============================================================================================================================== * Hadeel Gassim Hassan * Abeer Babiker Idris * Mohamed A. Hassan * Hisham N. Altayb * Kyakonye Yasin * Nazar Beirage * Muzamil M. Abdel Hamid ## Abstract **Background** There is an increase in the prevalence of *Helicobacter pylori* infection in Sudan, accompanied by a high incidence of upper gastrointestinal malignancy. The cytotoxin-associated gene *cagA* gene is a marker of a pathogenicity island (PAI) in *H. pylori* and plays a crucial role in determining the clinical outcome of *Helicobacter* infections. **Objective** This study aimed to determine the frequency and heterogeneity of the *cagA* gene of *H. pylori* and correlate the presence of *cagA* gene with clinical outcomes. **Materials and methods** Fifty endoscopy biopsies were collected from Fedail and Soba hospitals in Khartoum state. DNA was extracted using the Guanidine chloride method followed by PCR to amplify *16S rRNA* and *cagA* gene of *H. pylori* using specific primers. DNA amplicons of *cagA* gene were purified and sequenced. Bioinformatics and statistical analysis were done to characterize and to test the association between *cagA* gene and gastric complications. **Results** *CagA* gene was detected in 20/37(54%) of the samples that were found positive for *H. pylori*. There was no association between endoscopy finding and the presence of the *cagA* gene (p = 0.225). Specific amino acid variations were found at seven loci related to strains from a patient with duodenitis, gastric ulcer, and gastric atrophy (R448H, T457K, S460L, IT463-464VA, D470E, A482Q, KNV490-491-492TKT) while mutations in cancerous strain were A439P, T457P, and H500Y. **Conclusion** Disease-specific variations of *cagA* of *H. pylori* strains, in the region of amino acid residues 428-510, were evident among Sudanese patients with different gastroduodenal diseases. A novel mutation (K458N) was detected in a patient with duodenitis, which affects the positive electrostatic surface of *cagA*. Phylogenetic analysis showed a high level of diversity of *cagA* from Sudanese *H. pylori* strains. Keywords * *H.pylori* * *cagA* gene diversity * Sudan * virulence factors * *Insilco* ## 1. Introduction *Helicobacter pylori (H. pylori)* a bacterium is responsible for various gastrointestinal diseases, including gastritis, peptic ulcers, frequent bleeding events, gastric mucosa-associated lymphoid tissue (MALT) lymphoma, and carcinoma. (1, 2). A majority of infected individuals with *H. pylori* are asymptomatic(3, 4)and despite treatment, some infected patients may not clear the infection, thus leading to chronic complication. The possible reason for the various outcomes of *H. pylori* infection may relate to the variations in virulence factors of *H. pylori* strains in addition to host, environment, and dietary factors. (5) The cytotoxic associated gene A (*cagA*) is one of the most virulence factors which has been implicated as a possible marker to distinguish variations in the virulence of H. pylori strains. (6, 7)The *cagA* gene encodes a 120-145kDa protein, and it is located within the ∼40kb pathogenicity island (*cag* PAIs). (5) The *cag* PAIs expresses a type IV secretion systems (T4SS) which introduces the cagA effector protein into the host target cells where it undergoes tyrosine phosphorylation by Src and Ab1 kinases(8). The *cagA* protein is actively involved in the regulation of the spreading, migration, and adhesion of cells known to play a decisive role in mutagenic signal transduction, and stimulation of phosphatase activity also it induces vacuolation of primary human mucosal epithelial cells. (9, 10) *CagA* is the most intensely studied since its first association with the risk of gastric cancer. (10) Various studies have documented that infection with *H. pylori* strains expressing *cagA* was correlated with the development of severe gastrointestinal diseases such as atrophic gastritis, peptic ulcer, and gastric carcinoma. (11-15) *H. pylori* infect about half of the world population, but only a subset of 1% to 2% of those infected individuals will develop gastric malignancies. (1) The prevalence in developed countries is low 25%, a contrast to developing countries where the incidence is very high up to 90% (4, 16, 17) and the infection occurring early in life and tending to persist for longtime unless treated. (18)There is an increased in the prevalence of *H. pylori* infection in Sudan accompanied with high incidence of upper gastrointestinal malignancy (13.2%) when compared to previous study reported by Elhadi *et al*.(19) Therefore, the aim of this study was to determine the frequency and heterogeneity of *cagA* and its correlation with severe gastroduodenal diseases among Sudanese patients. To our knowledge, this is the first study in Sudan that reports the association of *cagA* and its heterogeneity to clinical outcomes of *H. pylori* infection. The molecular characterization of *cagA* and its mutations in amino acid sequence 428-510 were analyzed by using *in silico* tools. According to the X-ray crystallographic analysis of the N-terminal *cagA* fragment (residues 1-876) of the reference strain (*H. pylori* strain 26695),(20) amino acid sequence 428-510is located in domain II which tethers CagA to the plasma membrane by interacting with membrane phosphatidylserine. It is characterized by a sizeable antiparallel β-sheet and an insertion of a subdomain (residues 370-446) between β5 and β8 strands. (21) ## 2. Materials and methods ### 2.1 Study design and study population A prospective cross -sectional hospital-based study was conducted in Fedial and Soba hospitals in Khartoum state between February and May 2016. Patients who referred to hospitals for upper gastrointestinal tract endoscopy and suspected to be infected with *H. pylori* were enrolled. Patients who had received nonsteroidal anti-inflammatory drugs and proton pump inhibitor (PPI) were excluded from the study, and none of the patients had recently been prescribed antibiotics. ### 2.2 Clinical Samples The ethical approval was granted by the Institute of Endemic Diseases. All study participants completed informed consent forms prior to the commencement in the study. A total of fifty patients were selected for the study, twenty-five of these patients were males, and twenty-five were females. Specimens were obtained from all patients and tentatively diagnosed by the gastroenterologist as having a helicobacter infection. The samples were labeled and transferred to the laboratory for DNA analysis. Patient endoscopy findings included gastritis, gastric ulcer, atrophy gastric, gastritis, and duodenitis. ### 2.3 Extraction of DNA and PCR analysis DNA was extracted using Guanidine chloride method 17. PCR amplification was done using specific primers for *H. pylori* 16S rRNA gene F:5’GCGACCTGCTGGAACATTAC-3’ and R:5’GCGTTAGCTGCATTACTGGAGA-3’. A ready master pre-mix (Maxime, iNtRON BIOTECHNOLOGY, Korea) was used. A total volume of 25µl contained a1μl of each primer (10 pmole),1μl of DNA template and 22μl of de-ionized sterile water. A PCR reaction was carried out using a thermocycler (SensoQuest, Germany) following a protocol described by William *et al*. (22)*CagA* gene was amplified using primer set F:5’ATAATGCTAAATTAGACAACTTGAGCGA-3’ R:5’TTAGAATAATCAACAAACATCACGCCA-3’ using a method defined previously. (23) PCR amplicons were analysed on 1.5% Agarose gel stained with 1µl ethidium bromide (10mg/ml). Electrophoresis was done at 90V and 25mA. DNA bands were estimated using a 100bp DNA ladder (iNtRON BIOTECHNOLOGY, Korea European Biotech). The bands were visualized using Gel documentation systemBioDoc-It UVP, Cambridge UK). DNA purification and sequencing was performed commercially at Macrogen Inc, Korea. ### 2.4 Statistical and Bioinformatics analysis Data were analyzed using IBM SPSS Statistical software (Statistical Package for the Social Sciences). Chi-square was used to test the association of gastric complications with *cagA* gene. The nucleotide sequences of the cagA gene were compared for genetic diversity using nucleotide BLAST ([http://blast.ncbi.nlm.nih.gov/Blast.cgi](http://blast.ncbi.nlm.nih.gov/Blast.cgi)). (24) from NCBI, Highly similar sequences were retrieved and subjected to multiple sequence alignment using Clustal Omega - EMBL-EBI. (25) For nucleotides translation, evolutionary analysis and building a phylogenetic tree, GeneMarks (26)and jalview(27) were used, respectively. Modeling of 3D structural Protein was done by sending the protein sequence of the reference strain (*H. pylori* strain 26695)(20) to raptor X online server. (28) Structural homology modeling of this reference strain (NP_207343) was done due to the existence of short disordered stretches, common in all crystal forms,(21, 29-31) which their electron densities are not observed in any crystal such as residues 479-488 that is located in our region (residues 425-510). The Predicted structure was analyzed by using UCSF Chimera (version 1,11,2). (32) The nucleotide sequences of the *cagA* genes were deposited in the GenBank database (National Center for Biotechnology Information; [https://www.ncbi.nlm.nih.gov/](https://www.ncbi.nlm.nih.gov/)), Accession number of the strains with their clinical outcomes are presented in Table 1. View this table: [Table 1.](http://medrxiv.org/content/early/2019/09/27/19007435/T1) Table 1. Shows the accession number of the stains with their clinical outcomes ## 3. Results ### 3.1 Endoscopy finding Endoscopic examination was performed on all patients (n=50). Abnormal endoscopic finding was observed in 42/50(84%) of the patients, out of this 18/50(36%) were diagnosed with gastritis, 3/50(6%) with duodenitis, 12/50(24%) with gastric ulcers, 2/50(4%) with atrophy gastric, and 7/50(14%) with gastritis duodenitis. ### 3.2 Positive biopsy for *H. pylori* (16S rRNA) Fifty (n=50) samples were tested for the presence of 16S rRNA, 37/50(74%) were found to be positive, and 13/50(26%) were found to be negative for *H. pylori*. (Figure 1). ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2019/09/27/19007435/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2019/09/27/19007435/F1) Figure 1: Amplified *H. pylori*16SrRNAgene PCR products from endoscopy biopsy. Lane 1: negative control (1), lanes (2,4,5) is16srRNAof *H*.*pylori* positive (168pb); MW: 100 bp DNA molecular weight marker ### 3.3 Frequency of *cagA* gene The *cagA* gene frequency was found to be 20/37 (54%) among positive samples for (16S rRNA) of *H. pylori*. In gastric atrophy patients, it was found to be 2/20(10%),4/20(20%) in normal findings endoscopy patients,6/20(30%) in gastric ulcer patients,2/20(10%) in duodenitis patients, and gastritis patients were found to be 4/20(25%). (Table 2). View this table: [Table 2.](http://medrxiv.org/content/early/2019/09/27/19007435/T2) Table 2. Frequency of *cagA* gene in *H. pylori* strains from various gastroduodenal disease. ### 3.4 Variations of amino acid among *cagA* protein In comparison with strain 26695 (reference strain), disease-specific variations at different *cagA* loci were detected at a region in domain II (amino acid sequence 428-510) as illustrated (Figure2 and Figure 4).The mutations in amino acids R448H, T457K, S460L, IT463-464VA, D470E, A482Q, and KNV490-491-492TKT were found in *cagA* of duodenitis, gastric ulcer and gastric atrophy caused strains while mutations in cancerous strain were A439P, T457P, and H500Y. The amino acids at 467 and 497 loci were mutated in all strains regardless of gastrointestinal diseases. ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2019/09/27/19007435/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2019/09/27/19007435/F2) Figure 2: a) PCR gel showing bands corresponding to the *H. pylori cagA* gene from endoscopy biopsy. Lane 1: negative control (1), lanes 2,3,5 are Cag A positive genes of *H. pylori* (298pb), lane 4 negative sample.MW: 100bpDNA molecular weight marker. b) Shows the sequence chromatogram of codon 458 in normal finding and duodenitis causing strains, respectively by using Finch TV. c) Multiple sequence alignment of *cagA* gene sequences with other selected global strains obtained from GenBank. The different loci are shown in black letters, and white dots indicate similarity. The red boxes highlight amino acid variations at seven loci that are related to gastric ulcer, duodenitis, and gastric atrophy caused strains. Sudan-11 strain represents a cancerous strain. Secondary structure referred to strain 26695 (21). ![Figure 4:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2019/09/27/19007435/F3.medium.gif) [Figure 4:](http://medrxiv.org/content/early/2019/09/27/19007435/F3) Figure 4: a) Structural homology modeling of *cagA* derived from *H. pylori* strain 26695(20). b) and c) Shows loci of specific-disease mutations observed in duodenitis, ulcer, and atrophy gastric strains. The cyan color represents the locus of the significant novel amino acid substitution (458K) in duodenitis strain. ### 3.5 Phylogenetic analysis Phylogenetic analysis of *cagA* with global *H. pylori* strains revealed that the Sudanese *H*.*pylori* strains were divided into two major groups. One group involves strains that cause duodenitis, gastric ulcer, and gastric atrophy while cancerous strain belongs to another common ancestor, as illustrated in Figure5. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2019/09/27/19007435/F4.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2019/09/27/19007435/F4) Figure 2. The neighbor-joining tree for the *Helicobacter pylori cagA* gene of 16 Sudanese strains compared with 20 global reference strains from the Gen Bank database. The Sudanese strains are divided into two major groups Strains that cause duodenitis, gastric ulcer and gastric atrophy are colored in red while the blue color represents cancerous strain. ## 4. Discussion In this study, the analysis of amino acid sequence 425-510 of *cagA* indicated that the mutations at this region are distinguishable between gastrointestinal diseases. This indication is in agreement with the result obtained by Sung Soo *et al*., *2015* which suggested that the mutations of *cagA* of *H. pylori*, especially in C-terminus, are distinguishable between duodenal ulcer and gastric carcinoma. (33) In Sudan-9 strain that caused duodenitis, a novel mutation in lysine residue at 458 which constitutes with other lysine residues located in β9, β10 and α18; and two arginine residues (R624 and R626) the basic amino-acid cluster. (21) The basic amino-acid cluster provides a positive electrostatic surface potential, which is crucial for the CagA and phospholipid of the plasma membrane interaction. This interaction is essential for the delivery of CagA into host cells and also for the localization of delivered CagA to the inner face of the plasma membrane in epithelial cells. (34) Specific amino acid variations were found at seven loci (R448H, T457K, S460L, IT463-464VA, D470E, A482Q, KNV490-491-492TKT) which were related to strains detected in patients with gastric atrophy, gastric ulcer, and duodenitis. These mutations were also detected in Japanese and American strains isolated from patients with atrophy (Oki120, Oki112, Oki128, and Ok139 strains) (35, 36) and duodenal ulcer (Oki898 and Ok204 and J99 strains). (35-37) However, there are some strains of *H. pylori* implicated in both diseases gastric and duodenal ulcer (e.g., Germany strain UH44);(38) and maybe Sudanese strains are one of them. Two strains (Sudan-1 and Sudan-3) were detected from patients with normal gastroduodenal finding had the same mutations of gastric ulcer strains. While the third one (Sudan-2) and gastritis strains share the same mutations of cancerous strain (A439P, T457P, N468K, and H500Y).The infection of those patients could be a new type and need more time to reflect the complications or could be due to host/environmental factors that delay the appearance of complications. (39-41) This finding is partially in agreement with the result obtained by Masahiko *et al*., that the Amino acid frequency of gastritis samples seems to be intermediate between gastric cancer and MALT lymphoma samples. (42) In contrast, two Japanese strains (NCTC11637 and NCTC11638) isolated from patients with gastritis do not have any mutations in this region (amino acid residues 428-510) except N467G which was found in all our sequences in regardless to gastroduodenal diseases.(43) In this study, the frequency of *H. pylori* infection among endoscopics was found to be 37/50 (74%) using PCR; this result was higher compared to the prevalence of 18/81 (22.2%) reported by Mona *et al*., 2015 had been used culture methods. (44) Recently, assays based on PCR have been used to detect *H. pylori* DNA and its virulence genes in gastric biopsies with high specificity and quick results have been obtained without the requirement for special transport conditions. However, the presence of the *cagA* gene has been confirmed in 54% of the Sudanese *H. pylori* strains. The frequency of *cagA* positive *H. pylori* varies from one geographic region to another. In Sri Lanka is 48%, in the Netherlands is 67%, in Germany is 72%, in the United States is 81%, in Nigeria is 93% and in Korea is 97%. (2) The association between the genotype of *cagA* and clinical outcome was examined. There was no significant association between the genotype of *cagA* and clinical outcome (X2=1.567a; df=1; p=0.225). This finding agrees with the study conducted by Wen *et*.*al, 2004* in East Asia,(45) and disagrees with a study that was done in Chinese population that showing 95% positive for *cagA gene* among patients with peptic ulcer disease and 100% with chronic gastritis.(46)This is could be related to geographic strain distribution ;therefore more studies should be considered to include more sample size include ## Conclusion Disease-specific variation of *CagA* of *H. pylori* strains, in the region of amino acid residues 428-510, are found among Sudanese patients. A novel mutation (K458N) was detected in the basic amino-acid cluster, which provides a positive electrostatic surface for *CagA*. Phylogenetic analysis showed a high level of diversity of *cagA* among Sudanese *H. pylori* strains. ## Data Availability All important data are available in the manuscript ## Funding This study was funded by TWAS research grant No: 17-516 RG/BIO/AF/AC_G-FR3240297732 ## Conflicts of Interest The authors declare that there is no conflict of interest in this research work. ## Acknowledgements * Received September 21, 2019. * Revision received September 21, 2019. * Accepted September 27, 2019. * © 2019, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NoDerivs 4.0 International), CC BY-ND 4.0, as described at [http://creativecommons.org/licenses/by-nd/4.0/](http://creativecommons.org/licenses/by-nd/4.0/) ## References 1. 1.Testerman TL, Morris J. Beyond the stomach: an updated view of Helicobacter pylori pathogenesis, diagnosis, and treatment. World J Gastroenterol. 2014;20(36):12781–808. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3748/wjg.v20.i36.12781&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25278678&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) 2. 2.Bindayna KM, Al Baker WA, Botta GA. Detection of Helicobacter pylori cagA gene in gastric biopsies, clinical isolates, and faeces. Indian J Med Microbiol. 2006;24(3):195–200. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16912439&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) 3. 3.Abdallah TM, Mohammed HB, Mohammed MH, Ali AAA. Sero-prevalence and factors associated with Helicobacter pylori infection in Eastern Sudan. Asian Pacific Journal of Tropical Disease. 2014;4(2):115–9. 4. 4.Afihene MKY, Denyer M, Amuasi JH, Boakye I, Nkrumah K. Prevalence of Helicobacter pylori and Endoscopic findings among dyspeptics in Kumasi, Ghana. Open Science Journal of Clinical Medicine. 2014;2(3): 63–8. 5. 5.Matsunari O, Shiota S, Suzuki R, Watada M, Kinjo N, Murakami K, et al. Association between Helicobacter pylori virulence factors and gastroduodenal diseases in Okinawa, Japan. Journal of clinical microbiology. 2012;50(3):876–83. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNtIjtzOjU6InJlc2lkIjtzOjg6IjUwLzMvODc2IjtzOjQ6ImF0b20iO3M6Mzk6Ii9tZWRyeGl2L2Vhcmx5LzIwMTkvMDkvMjcvMTkwMDc0MzUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 6. 6.Hoshino FB, Katayama K, Watanabe K, Takahashi S, Uchimura H, T. A. Heterogeneity found in the cagA gene of Helicobacter pylori from Japanese and non-Japanese isolates. J Gastroenterol. 2000;35(12):890–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s005350070002&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11573724&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) 7. 7.Crabtree JE, Covacci A, Farmery SM, Xiang Z, Tompkins DS, Perry S, et al. Helicobacter pylori induced interleukin-8 expression in gastric epithelial cells is associated with CagA positive phenotype. Journal of clinical pathology. 1995;48(1):41–5. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToiamNsaW5wYXRoIjtzOjU6InJlc2lkIjtzOjc6IjQ4LzEvNDEiO3M6NDoiYXRvbSI7czozOToiL21lZHJ4aXYvZWFybHkvMjAxOS8wOS8yNy8xOTAwNzQzNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 8. 8.Salama N, Guillemin K, McDaniel TK, Sherlock G, Tompkins L, Falkow S. A whole-genome microarray reveals genetic diversity among Helicobacter pylori strains. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(26):14668–73. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiOTcvMjYvMTQ2NjgiO3M6NDoiYXRvbSI7czozOToiL21lZHJ4aXYvZWFybHkvMjAxOS8wOS8yNy8xOTAwNzQzNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 9. 9.Kim JM, Kim JS, Jung HC, Song IS, Kim CY. Virulence factors of Helicobacter pylori in Korean isolates do not influence proinflammatory cytokine gene expression and apoptosis in human gastric epithelial cells, nor do these factors influence the clinical outcome. Journal of gastroenterology. 2000;35(12):898–906. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s005350070003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11573725&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000166041700003&link_type=ISI) 10. 10.Olbermann P, Josenhans C, Moodley Y, Uhr M, Stamer C, Vauterin M, et al. A global overview of the genetic and functional diversity in the Helicobacter pylori cag pathogenicity island. PLoS genetics. 2010;6(8):e1001069–e. 11. 11.Peek RM, Jr.., Miller GG, Tham KT, Perez-Perez GI, Zhao X, Atherton JC, et al. Heightened inflammatory response and cytokine expression in vivo to cagA+ Helicobacter pylori strains. Laboratory investigation; a journal of technical methods and pathology. 1995;73(6):760–70. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8558837&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1995TL72500004&link_type=ISI) 12. 12.Navaglia F BD, Piva MG, Brigato L, Stefani A, Dal Bò N, Di Mario F, Rugge M, Plebani M. Helicobacter pylori cytotoxic genotype is associated with peptic ulcer and influences serology. The American journal of gastroenterology. 1998;93(2):227–30. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1572-0241.1998.00227.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9468248&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000072477100022&link_type=ISI) 13. 13.Gunn MC, Stephens JC, Stewart JA, Rathbone BJ, West KP. The significance of cagA and vacA subtypes of Helicobacter pylori in the pathogenesis of inflammation and peptic ulceration. Journal of clinical pathology. 1998;51(10):761–4. [Abstract](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToiamNsaW5wYXRoIjtzOjU6InJlc2lkIjtzOjk6IjUxLzEwLzc2MSI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzA5LzI3LzE5MDA3NDM1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 14. 14.Basso D, Navaglia F, Brigato L, Piva MG, Toma A, Greco E, et al. Analysis of Helicobacter pylori vacA and cagA genotypes and serum antibody profile in benign and malignant gastroduodenal diseases. Gut. 1998;43(2):182–6. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ3V0am5sIjtzOjU6InJlc2lkIjtzOjg6IjQzLzIvMTgyIjtzOjQ6ImF0b20iO3M6Mzk6Ii9tZWRyeGl2L2Vhcmx5LzIwMTkvMDkvMjcvMTkwMDc0MzUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 15. 15.Rudi J, Rudy A, Maiwald M, Kuck D, Sieg A, Stremmel W. Direct determination of Helicobacter pylori vacA genotypes and cagA gene in gastric biopsies and relationship to gastrointestinal diseases. The American journal of gastroenterology. 1999;94(6):1525–31. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1572-0241.1999.1138_a.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10364019&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) 16. 16.Holcombe C. Helicobacter pylori: the African enigma. Gut. 1992;33:429–31. [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo2OiJndXRqbmwiO3M6NToicmVzaWQiO3M6ODoiMzMvNC80MjkiO3M6NDoiYXRvbSI7czozOToiL21lZHJ4aXYvZWFybHkvMjAxOS8wOS8yNy8xOTAwNzQzNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 17. 17.Quek C, Pham ST, Tran KT, Pham BT, Huynh LV, Luu NB, et al. Antimicrobial susceptibility and clarithromycin resistance patterns of Helicobacter pylori clinical isolates in Vietnam. F1000Research. 2016;5:671. 18. 18.Rowland M, Drumm B. Clinical significance of Helicobacter infection in children. Br Med Bull. 1998;54(1):95–103. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9604435&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000075220800011&link_type=ISI) 19. 19.Aa E, Ho M, Th M, Os M, Ha E. Pattern of Endoscopic Findings of Upper Gastrointestinal Tract in Omdurman Teaching. Sudan JMS 2004;9(2):71–4. 20. 20.Xu S, Zhang C, Miao Y, Gao J, Xu D. Effector prediction in host-pathogen interaction based on a Markov model of a ubiquitous EPIYA motif. BMC genomics. 2010;11 Suppl 3(Suppl 3):S1–S. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1471-2164-11-S5-S1&link_type=DOI) 21. 21.Hayashi T, Senda M, Morohashi H, Higashi H, Horio M, Kashiba Y, et al. Tertiary structure-function analysis reveals the pathogenic signaling potentiation mechanism of Helicobacter pylori oncogenic effector CagA. Cell host & microbe. 2012;12(1):20–33. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.chom.2012.05.010&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22817985&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000306941900005&link_type=ISI) 22. 22.Gramley WA, Asghar A, Frierson HF, Jr.., Powell SM. Detection of Helicobacter pylori DNA in fecal samples from infected individuals. Journal of clinical microbiology. 1999;37(7):2236–40. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNtIjtzOjU6InJlc2lkIjtzOjk6IjM3LzcvMjIzNiI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzA5LzI3LzE5MDA3NDM1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 23. 23.Walencka M, Gonciarz W, Mnich E, Gajewski A, Stawerski P, Knapik-Dabrowicz A, et al. The microbiological, histological, immunological and molecular determinants of Helicobacter pylori infection in guinea pigs as a convenient animal model to study pathogenicity of these bacteria and the infection dependent immune response of the host. Acta biochimica Polonica. 2015;62(4):697–706. 24. 24.Altschul SF, Madden TL, Schäffer AA, J Zhang ZZ, Miller W, Lipman DJ. Gapped BLAST and PSI-BLAST. A new generation of protein database search programmes. Nucleic Acids Res. 1997;25(17):3389–402. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/25.17.3389&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9254694&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1997XU79300002&link_type=ISI) 25. 25.McWilliam H, Li W, Uludag M, Squizzato S, Park YM, Buso N, et al. Analysis Tool Web Services from the EMBL-EBI. Nucleic Acids Res. 2013;41(Web Server issue): W597-600. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkt376&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23671338&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000323603200095&link_type=ISI) 26. 26.Besemer J, Lomsadze A, Borodovsky M. GeneMarkS: a self-training method for prediction of gene starts in microbial genomes. Implications for finding sequence motifs in regulatory regions. Nucleic Acids Res. 2001;29(12):2607–18. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/29.12.2607&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11410670&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000169616100022&link_type=ISI) 27. 27.Waterhouse AM, Procter JB, Martin DMA, Clamp M, Barton GJ. Jalview Version 2--a multiple sequence alignment editor and analysis workbench. Bioinformatics (Oxford, England). 2009;25(9):1189–91. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btp033&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19151095&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000265523300016&link_type=ISI) 28. 28.Källberg M, Wang H, Wang S, Peng J, Wang Z, Lu H, et al. Template-based protein structure modeling using the RaptorX web server. Nature Protocols. 2012;7:1511. 29. 29.Nesic D, Miller MC, Quinkert ZT, Stein M, Chait BT, Stebbins CE. Helicobacter pylori CagA inhibits PAR1-MARK family kinases by mimicking host substrates. Nature structural & molecular biology. 2010;17(1):130–2. 30. 30.Kaplan-Turkoz B, Jimenez-Soto LF, Dian C, Ertl C, Remaut H, Louche A, et al. Structural insights into Helicobacter pylori oncoprotein CagA interaction with beta1 integrin. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(36):14640–5. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTA5LzM2LzE0NjQwIjtzOjQ6ImF0b20iO3M6Mzk6Ii9tZWRyeGl2L2Vhcmx5LzIwMTkvMDkvMjcvMTkwMDc0MzUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 31. 31.Nesic D, Buti L, Lu X, Stebbins CE. Structure of the Helicobacter pylori CagA oncoprotein bound to the human tumor suppressor ASPP2. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(4):1562–7. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiMTExLzQvMTU2MiI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzA5LzI3LzE5MDA3NDM1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 32. 32.Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, et al. UCSF Chimera--a visualization system for exploratory research and analysis. Journal of computational chemistry. 2004;25(13):1605–12. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/jcc.20084&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15264254&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000223379100005&link_type=ISI) 33. 33.Kim SS, Cheung DY, Park S-H. Differences of mutation of helicobacter pylori cagA gene isolated from duodenal ulcer and gastric cancer. Journal of Clinical Oncology. 2015;33(3_suppl):69-. 34. 34.Murata-Kamiya N, Kikuchi K, Hayashi T, Higashi H, Hatakeyama M. Helicobacter pylori exploit host membrane phosphatidylserine for delivery, localization, and pathophysiological action of the CagA oncoprotein. Cell host & microbe. 2010;7(5):399–411. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.chom.2010.04.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20478541&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000277954500010&link_type=ISI) 35. 35.Satou K, Shiroma A, Teruya K, Shimoji M, Nakano K, Juan A, et al. Complete Genome Sequences of Eight Helicobacter pylori Strains with Different Virulence Factor Genotypes and Methylation Profiles, Isolated from Patients with Diverse Gastrointestinal Diseases on Okinawa Island, Japan, Determined Using PacBio Single-Molecule Real-Time Technology. Genome announcements. 2014;2(2). 36. 36.Yamazaki S, Yamakawa A, Okuda T, Ohtani M, Suto H, Ito Y, et al. Distinct diversity of vacA, cagA, and cagE genes of Helicobacter pylori associated with peptic ulcer in Japan. Journal of clinical microbiology. 2005;43(8):3906–16. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNtIjtzOjU6InJlc2lkIjtzOjk6IjQzLzgvMzkwNiI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzA5LzI3LzE5MDA3NDM1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 37. 37.Beaulaurier J, Zhang XS, Zhu S, Sebra R, Rosenbluh C, Deikus G, et al. Single molecule-level detection and long read-based phasing of epigenetic variations in bacterial methylomes. Nature communications. 2015;6:7438. 38. 38.Backert S, Schwarz T, Miehlke S, Kirsch C, Sommer C, Kwok T, et al. Functional analysis of the cag pathogenicity island in Helicobacter pylori isolates from patients with gastritis, peptic ulcer, and gastric cancer. Infection and immunity. 2004;72(2):1043–56. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiaWFpIjtzOjU6InJlc2lkIjtzOjk6IjcyLzIvMTA0MyI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzA5LzI3LzE5MDA3NDM1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 39. 39.Testerman TL, Morris J. Beyond the stomach: An updated view of Helicobacter pylori pathogenesis, diagnosis, and treatment. World journal of gastroenterology. 2014;20(36):12781–808. 40. 40.D. G. Solving the African enigma: parasites may have their place. Gastroenterology. 2000;119(3):611. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11023356&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) 41. 41.Kidd M, Louw JA, Marks IN. Helicobacter pylori in Africa: Observations on an ‘enigma within an enigma’. Journal of Gastroenterology and Hepatology. 1999;14(9):851–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1046/j.1440-1746.1999.01975.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10535465&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) 42. 42.Hashinaga M, Suzuki R, Akada J, Matsumoto T, Kido Y, Okimoto T, et al. Differences in amino acid frequency in CagA and VacA sequences of Helicobacter pylori distinguish gastric cancer from gastric MALT lymphoma. Gut pathogens. 2016;8:54. 43. 43.Achtman M, Azuma T, Berg DE, Ito Y, Morelli G, Pan ZJ, et al. Recombination and clonal groupings within Helicobacter pylori from different geographical regions. Molecular microbiology. 1999;32(3):459–70. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1046/j.1365-2958.1999.01382.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10320570&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000080720900003&link_type=ISI) 44. 44.Mamoun M, M ES, Enan KA, Abdo AE, Hassan MA. Molecular Identification 0f 16s Ribosomal RNA Gene of Helicobacter pylori Isolated from Gastric Biopsies in Sudan. American Journal of Microbiological Research. 2015;3(2):50–4. 45. 45.Zhou W, Yamazaki S, Yamakawa A, Ohtani M, Ito Y, Keida Y, et al. The diversity of vacA and cagA genes of Helicobacter pylori in East Asia. FEMS immunology and medical microbiology. 2004;40(1):81–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0928-8244(03)00299-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=14734191&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F09%2F27%2F19007435.atom) 46. 46.Pan Z, Bart A, Feller M, Xiao S, Tytgat G, J. N. cagA -Positive Helicobacter pylori Populations in China and The Netherlands Are Distinct. Tailieuvn. 1998;66:1822–6