101
|
Oikawa T, Asano N, Imatani A, Ohyauchi M, Fushiya J, Kondo Y, Abe Y, Koike T, Iijima K, Shimosegawa T. Gene polymorphisms of NOD1 and interleukin-8 influence the susceptibility to erosive esophagitis in Helicobacter pylori infected Japanese population. Hum Immunol 2012; 73:1184-9. [PMID: 22902391 DOI: 10.1016/j.humimm.2012.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Revised: 08/09/2012] [Accepted: 08/09/2012] [Indexed: 12/25/2022]
Abstract
Helicobacter pylori (H. pylori) infection generally protects patients from erosive esophagitis through reduction of acid production due to gastric mucosal atrophy. However, there are H. pylori infected patients who still have erosive esophagitis. The reason for this discrepancy remains unclear. We have previously reported that polymorphisms in IL-8 promoter region influence the susceptibility of H. pylori related diseases. On the other hand, nucleotide-binding oligomerization domain 1 (NOD1) is known to play an important role in H. pylori infection. Hence, we hypothesized polymorphisms of these two molecules in H. pylori infected patients may influence the susceptibility to erosive esophagitis. Genomic DNA was extracted from 312 H. pylori infected Japanese, consisting of 110 patients with erosive esophagitis and 202 healthy controls. ND1+32656 T/GG and IL-8-251 A/T polymorphisms were genotyped by direct sequencing. ND1+32656 GG allele and IL-8-251 T/T allele increased the risk of erosive esophagitis with odds ratio (OR) of 1.9 (95% confidence interval (CI) 1.1-3.0, p=0.013) and 1.7 (95% CI 1.1-2.8, p=0.036), respectively. Combination of these two alleles increased the risk with OR of 3.2(95% CI 1.6-6.5, p=0.001). In conclusion, ND1+32656 GG and IL-8-251 T/T allele may be associated with less reactivity to H. pylori infection, and may increase the risk of erosive esophagitis even in H. pylori infected Japanese population.
Collapse
Affiliation(s)
- Tomoyuki Oikawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Pathak AK, Pelensky C, Boag B, Cattadori IM. Immuno-epidemiology of chronic bacterial and helminth co-infections: observations from the field and evidence from the laboratory. Int J Parasitol 2012; 42:647-55. [PMID: 22584129 DOI: 10.1016/j.ijpara.2012.04.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 03/21/2012] [Accepted: 04/12/2012] [Indexed: 12/12/2022]
Abstract
Co-infections can alter the host immune responses and modify the intensity and dynamics of concurrent parasitic species. The extent of this effect depends on the properties of the system and the mechanisms of host-parasite and parasite-parasite interactions. We examined the immuno-epidemiology of a chronic co-infection to reveal the immune mediated relationships between two parasites colonising independent organs, and the within-host molecular processes influencing the dynamics of infection at the host population level. The respiratory bacterium, Bordetella bronchiseptica, and the gastrointestinal helminth, Graphidium strigosum, were studied in the European rabbit (Oryctolagus cuniculus), using long-term field data and a laboratory experiment. We found that 65% of the rabbit population was co-infected with the two parasites; prevalence and intensity of co-infection increased with rabbit age and exhibited a strong seasonal pattern with the lowest values recorded during host breeding (from April to July) and the highest in the winter months. Laboratory infections showed no significant immune-mediated effects of the helminth on bacterial intensity in the lower respiratory tract but a higher abundance was observed in the nasal cavity during the chronic phase of the infection, compared with single bacterial infections. In contrast, B. bronchiseptica enhanced helminth intensity and this was consistent throughout the 4-month trial. These patterns were associated with changes in the immune profiles between singly and co-infected individuals for both parasites. This study confirmed the general observation that co-infections alter the host immune responses but also highlighted the often ignored role of bacterial infection in helminth dynamics. Additionally, we showed that G. strigosum had contrasting effects on B. bronchiseptica colonising different parts of the respiratory tract. At the host population level our findings suggest that B. bronchiseptica facilitates G. strigosum infection, and re-infection with G. strigosum assists in maintaining bacterial infection in the upper respiratory tract and thus long-term persistence.
Collapse
Affiliation(s)
- Ashutosh K Pathak
- Center for Infectious Disease Dynamics and Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | |
Collapse
|
103
|
Ek C, Whary MT, Ihrig M, Bravo LE, Correa P, Fox JG. Serologic evidence that ascaris and toxoplasma infections impact inflammatory responses to Helicobacter pylori in Colombians. Helicobacter 2012; 17:107-15. [PMID: 22404440 PMCID: PMC3305284 DOI: 10.1111/j.1523-5378.2011.00916.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Helicobacter pylori-infected children from coastal Tumaco, Colombia, have more parasitism, and adults have lower gastric cancer risk compared with high-altitude Pasto/Tuquerres residents. Because helminth and Toxoplasma gondii infections alter helicobacter gastritis in rodent models, we determined whether seropositivity to Ascaris lumbricoides or T. gondii was associated with Th2-IgG1 or Th1-IgG2 responses to H. pylori. METHODS Sera (240) from the two populations were evaluated for A. lumbricoides and T. gondii seropositivity and results correlated with IgE and IgG isotype responses to H. pylori. RESULTS Most Tumaco children and adults were seropositive for A. lumbricoides (89%, 66%), T. gondii (59%, 98%), or both (45%, 66%). In contrast, seropositivity among Pasto/Tuquerres children was much lower (9%A. lumbricoides, 11%T. gondii, and 2% dual positive) but increased in adults (58%A. lumbricoides, 82%T. gondii, and 41% dual positive). A. lumbricoides seropositivity correlated with elevated IgE and anti-inflammatory Th2-IgG1 responses to H. pylori, while T. gondiigondii seropositivity was linked to elevated IgE, pro-inflammatory Th1-IgG2, IgG3, and IgG4 responses to H. pylori. Individuals with high T. gondii titers had reduced Th1-IgG2, IgG3, and IgG4 responses to H. pylori. CONCLUSIONS Results support regional differences for childhood parasitism and indicate A. lumbricoides and T. gondii infections may impact inflammatory responses to H. pylori and partially explain differences in gastric cancer risk in Colombia.
Collapse
Affiliation(s)
- Courtney Ek
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Mark T. Whary
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Melanie Ihrig
- Comparative Medicine Program, The Methodist Hospital Research Institute, Houston, Texas 77030, USA
| | - Luis E. Bravo
- Department of Pathology, Universidad del Valle School of Medicine, Cali, 25360, Colombia
| | - Pelayo Correa
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37240, USA
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
104
|
Quante M, Bhagat G, Abrams J, Marache F, Good P, Lee MD, Lee Y, Friedman R, Asfaha S, Dubeykovskaya Z, Mahmood U, Figueiredo JL, Kitajewski J, Shawber C, Lightdale C, Rustgi AK, Wang TC. Bile acid and inflammation activate gastric cardia stem cells in a mouse model of Barrett-like metaplasia. Cancer Cell 2012; 21:36-51. [PMID: 22264787 PMCID: PMC3266546 DOI: 10.1016/j.ccr.2011.12.004] [Citation(s) in RCA: 369] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 06/02/2011] [Accepted: 12/01/2011] [Indexed: 02/06/2023]
Abstract
Esophageal adenocarcinoma (EAC) arises from Barrett esophagus (BE), intestinal-like columnar metaplasia linked to reflux esophagitis. In a transgenic mouse model of BE, esophageal overexpression of interleukin-1β phenocopies human pathology with evolution of esophagitis, Barrett-like metaplasia and EAC. Histopathology and gene signatures closely resembled human BE, with upregulation of TFF2, Bmp4, Cdx2, Notch1, and IL-6. The development of BE and EAC was accelerated by exposure to bile acids and/or nitrosamines, and inhibited by IL-6 deficiency. Lgr5(+) gastric cardia stem cells present in BE were able to lineage trace the early BE lesion. Our data suggest that BE and EAC arise from gastric progenitors due to a tumor-promoting IL-1β-IL-6 signaling cascade and Dll1-dependent Notch signaling.
Collapse
Affiliation(s)
- Michael Quante
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München
- Corresponding authors: Timothy C. Wang, M.D., Division of Digestive and Liver Diseases, Columbia University Medical Center, 1130 St. Nicholas Avenue, Room 925, 9th Floor; New York, NY 10032, Phone: (212) 851-4581; Fax: (212) 851-4590; . Michael Quante, M.D., II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München, Phone: +49 89 4140 6795; Fax: +49 89 4140 6796;
| | - Govind Bhagat
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY
| | - Julian Abrams
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Frederic Marache
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Pamela Good
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Michele D. Lee
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Yoomi Lee
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Richard Friedman
- Department of Biomedical Informatics, Columbia University Medical Center, New York, NY
| | - Samuel Asfaha
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Zinaida Dubeykovskaya
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Umar Mahmood
- Nuclear Medicine & Molecular Imaging, Harvard Medical School and Massachusetts General Hospital, Boston, MA
| | - Jose-Luiz Figueiredo
- Center for Systems Biology, Harvard Medical School and Massachusetts General Hospital, Boston, MA
| | - Jan Kitajewski
- Pathology, Obstetrics and Gynecology, and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Carrie Shawber
- Pathology, Obstetrics and Gynecology, and Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Charles Lightdale
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
| | - Anil K. Rustgi
- Division of Gastroenterology, Department of Medicine and Genetics, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Irving Cancer Research Center, Department of Medicine, Columbia University Medical Center, New York, NY
- Corresponding authors: Timothy C. Wang, M.D., Division of Digestive and Liver Diseases, Columbia University Medical Center, 1130 St. Nicholas Avenue, Room 925, 9th Floor; New York, NY 10032, Phone: (212) 851-4581; Fax: (212) 851-4590; . Michael Quante, M.D., II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 München, Phone: +49 89 4140 6795; Fax: +49 89 4140 6796;
| |
Collapse
|
105
|
Thakar J, Pathak AK, Murphy L, Albert R, Cattadori IM. Network model of immune responses reveals key effectors to single and co-infection dynamics by a respiratory bacterium and a gastrointestinal helminth. PLoS Comput Biol 2012; 8:e1002345. [PMID: 22253585 PMCID: PMC3257297 DOI: 10.1371/journal.pcbi.1002345] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 11/25/2011] [Indexed: 12/22/2022] Open
Abstract
Co-infections alter the host immune response but how the systemic and local processes at the site of infection interact is still unclear. The majority of studies on co-infections concentrate on one of the infecting species, an immune function or group of cells and often focus on the initial phase of the infection. Here, we used a combination of experiments and mathematical modelling to investigate the network of immune responses against single and co-infections with the respiratory bacterium Bordetella bronchiseptica and the gastrointestinal helminth Trichostrongylus retortaeformis. Our goal was to identify representative mediators and functions that could capture the essence of the host immune response as a whole, and to assess how their relative contribution dynamically changed over time and between single and co-infected individuals. Network-based discrete dynamic models of single infections were built using current knowledge of bacterial and helminth immunology; the two single infection models were combined into a co-infection model that was then verified by our empirical findings. Simulations showed that a T helper cell mediated antibody and neutrophil response led to phagocytosis and clearance of B. bronchiseptica from the lungs. This was consistent in single and co-infection with no significant delay induced by the helminth. In contrast, T. retortaeformis intensity decreased faster when co-infected with the bacterium. Simulations suggested that the robust recruitment of neutrophils in the co-infection, added to the activation of IgG and eosinophil driven reduction of larvae, which also played an important role in single infection, contributed to this fast clearance. Perturbation analysis of the models, through the knockout of individual nodes (immune cells), identified the cells critical to parasite persistence and clearance both in single and co-infections. Our integrated approach captured the within-host immuno-dynamics of bacteria-helminth infection and identified key components that can be crucial for explaining individual variability between single and co-infections in natural populations. Infections with different infecting agents can alter the immune response against any one parasite and the relative abundance and persistence of the infections within the host. This is because the immune system is not compartmentalized but acts as a whole to allow the host to maintain control of the infections as well as repair damaged tissues and avoid immuno-pathology. There is no comprehensive understanding of the immune responses during co-infections and of how systemic and local mechanisms interact. Here we integrated experimental data with mathematical modelling to describe the network of immune responses of single and co-infection by a respiratory bacterium and a gastrointestinal helminth. We were able to identify key cells and functions responsible for clearing or reducing both parasites and showed that some mechanisms differed between type of infection as a result of different signal outputs and cells contributing to the immune processes. This study highlights the importance of understanding the immuno-dynamics of co-infection as a host response, how immune mechanisms differ from single infections and how they may alter parasite persistence, impact and abundance.
Collapse
Affiliation(s)
- Juilee Thakar
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Ashutosh K. Pathak
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Lisa Murphy
- Division of Animal Production and Public Health, Veterinary School, University of Glasgow, Glasgow, United Kingdom
| | - Réka Albert
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Physics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Isabella M. Cattadori
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
106
|
Gray BM, Eaton KA. Necropsy, blood, tissue collection, and mRNA isolation for detection of host cytokine gene expression. Methods Mol Biol 2012; 921:119-129. [PMID: 23015500 DOI: 10.1007/978-1-62703-005-2_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Processing of tissue and blood must be done in a systematic and controlled fashion in order to optimize results and allow comparison of samples between experiments and between laboratories. Here we present our protocols for blood and tissue processing.
Collapse
Affiliation(s)
- Brian M Gray
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | |
Collapse
|
107
|
Rogers AB. Gastric Helicobacter spp. in animal models: pathogenesis and modulation by extragastric coinfections. Methods Mol Biol 2012; 921:175-188. [PMID: 23015504 DOI: 10.1007/978-1-62703-005-2_21] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Animal models are used to study complex host, microbial, and environmental influences associated with gastric Helicobacter infection. Evidence that gastric helicobacters are pathogenic in animals first came from ferrets. Felids, nonhuman primates, and many other species also harbor stomach helicobacters. Today, mice are preferred by most researchers for scientific investigation because of cost-efficiencies, rapid reproduction, choice of laboratory reagents, and availability of genetically engineered models. Infection with Helicobacter felis or H. pylori Sydney strain-1 in appropriate mouse strains produces disease with remarkable similarities to H. pylori in humans. Due to recent advances in genetic engineering, in vivo imaging, and system-wide genomics and proteomics, these models will become even more widespread in the future. Recently, it has been shown that extragastric infections can dramatically affect the severity of disease induced by gastric Helicobacter spp. through heterologous immunity. These models provide proof-of-principle for the "African enigma" wherein gastric cancer is underrepresented in low-lying tropical countries with concurrently high H. pylori and internal parasite prevalence. Helicobacter gastritis and carcinogenesis in mouse models may be augmented or ameliorated by other infectious agents depending on the character of the invoked immune response. Knowledge gained from the Human Microbiome Project and other investigations is certain to shed new light on the influence of extragastric bacterial, viral, fungal, and parasitic coinfections on H. pylori-associated peptic ulcer disease and gastric adenocarcinoma.
Collapse
Affiliation(s)
- Arlin B Rogers
- Lineberger Comprehensive Cancer Center and Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
108
|
Abstract
Animal models are essential for in vivo analysis of Helicobacter-related diseases. Transgenic mice and Mongolian gerbil models have been the corner stone of present research focusing on both bacterial virulence factors and host response to infection. Establishing a reproducible rodent model of persistent Helicobacter pylori infection that resembles the H. pylori-associated gastritis observed in humans was a considerable challenge until Lee et al. (Gastroenterology 112:1386-1397, 1997) successfully adapted a clinical Cag A- and Vac A-expressing strain for the mouse stomach. This so-called SS1 (Sydney) strain has since been extensively used for H. pylori research; other rodent-adapted Helicobacter strains have subsequently been developed and utilized in wild-type and genetically engineered rodent models. These bacteria include both H. pylori and the larger but related species H. felis (originally isolated from cats). In this chapter we focus mainly on these two Helicobacter strains and review the rodent models that have been employed to investigate how Helicobacter species induce gastric inflammation and disease.
Collapse
|
109
|
Houghton JM. Helicobacter species methods and protocols. Introduction. Methods Mol Biol 2012; 921:1-5. [PMID: 23015484 DOI: 10.1007/978-1-62703-005-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Helicobacter infection is a chronic persistent condition which is responsible for the majority of cases of gastric and duodenal ulcers, and gastric cancer. The study of the bacteria, the interaction of the bacteria with the host, and the host immune response has greatly benefited from standardization of culture techniques and animal models. The following chapters will describe the clinical aspects of infection and touch on the important techniques for optimal investigation of this infection.
Collapse
Affiliation(s)
- Jean Marie Houghton
- Division of Gastroenterology, Department of Medicine, University of Massachusetts Medical School Worcester, Worcester, MA, USA.
| |
Collapse
|
110
|
Immunological response to parenteral vaccination with recombinant hepatitis B virus surface antigen virus-like particles expressing Helicobacter pylori KatA epitopes in a murine H. pylori challenge model. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 19:268-76. [PMID: 22205658 DOI: 10.1128/cvi.05295-11] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Virus-like particles (VLPs) based on the small envelope protein of hepatitis B virus (HBsAg-S) are immunogenic at the B- and T-cell level. In this study, we inserted overlapping sequences encoding the carboxy terminus of the Helicobacter pylori katA gene product into HBsAg-S. The HBsAg-S-KatA fusion proteins were able to assemble into secretion-competent VLPs (VLP-KatA). The VLP-KatA proteins were able to induce KatA-specific antibodies in immunized mice. The mean total IgG antibody titers 41 days post-primary immunization with VLP-KatA (2.3 × 10(3)) were significantly greater (P < 0.05) than those observed for vaccination with VLP alone (5.2 × 10(2)). Measurement of IgG isotypes revealed responses to both IgG1 and IgG2a (mean titers, 9.0 × 10(4) and 2.6 × 10(4), respectively), with the IgG2a response to vaccination with VLP-KatA being significantly higher than that for mice immunized with KatA alone (P < 0.05). Following challenge of mice with H. pylori, a significantly reduced bacterial load in the gastric mucosa was observed (P < 0.05). This is the first report describing the use of VLPs as a delivery vehicle for H. pylori antigens.
Collapse
|
111
|
Pathak AK, Biarnes MC, Murphy L, Cattadori IM. Snapshot of spatio-temporal cytokine responses to single and co-infections with helminths and bacteria. RESULTS IN IMMUNOLOGY 2011; 1:95-102. [PMID: 24371558 DOI: 10.1016/j.rinim.2011.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 10/21/2011] [Accepted: 10/31/2011] [Indexed: 12/24/2022]
Abstract
Cytokines play a key role in maintaining communication between organs and in so doing modulate the interaction between concurrent infections. The extent of these effects depends on the properties of the organ infected and the intensity and type of infections. To determine systemic bystander effects among organs, IFN-γ, IL-4 and IL-10 gene expression was quantified at 7 days post-challenge in directly infected and uninfected organs during single and co-infections with the respiratory bacterium Bordetella bronchiseptica and the gastrointestinal helminths Graphidium strigosum and Trichostrongylus retortaeformis. Results showed that cytokine expression in a specific organ was influenced by the type of infection occurring in another organ, and this bystander effect was more apparent in some organs than others. Within the same organ the relative cytokine expression was consistent across infections, although some cytokines were more affected by bystander effects than others. For the infected gastrointestinal tract, a stronger cytokine response was observed in the tissue that harbored the majority of helminths (i.e. duodenum and fundus). Overall, co-infections altered the intensity but to a lesser extent the relative cytokine profile against the focal infection, indicating clear bystander effects and low organ compartmentalization. However, organs appear to actively modulate cytokine expression to avoid potential immuno-pathological consequences.
Collapse
Key Words
- AR-1, autoregressive function of order 1
- B, B. bronchiseptica single infection
- BG, B. bronchiseptica+G. strigosum dual-infection
- BT, B. bronchiseptica+T. retortaeformis dual-infection
- BTG, B. bronchiseptica+T. retortaeformis+G. strigosum triple infection
- Bordetella bronchiseptica
- Bystander effects
- Co-infections
- Cytokine gene expression
- DPI, days post-infection
- GLM, generalized linear models
- Graphidium strigosum
- IFN-γ, Interferon-gamma
- IL-10, Interleukin-10
- IL-4, Interleukin-4
- LME-REML, linear mixed effect models with restricted maximum likelihood
- SI, small intestine
- T, T. retortaeformis single infection
- TG, T. retortaeformis+G. strigosum dual helminth co-infection
- Trichostrongylus retortaeformis
Collapse
Affiliation(s)
- Ashutosh K Pathak
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802, USA ; Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Michael C Biarnes
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Lisa Murphy
- Division of Animal Production and Public Health, Veterinary School, University of Glasgow, Glasgow G61 1QH, UK
| | - Isabella M Cattadori
- Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802, USA ; Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
112
|
González CA, Agudo A. Carcinogenesis, prevention and early detection of gastric cancer: where we are and where we should go. Int J Cancer 2011; 130:745-53. [PMID: 21918974 DOI: 10.1002/ijc.26430] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 09/01/2011] [Indexed: 12/15/2022]
Abstract
Helicobacter pylori is the most common cause of gastric cancer (GC), though smoking, alcohol, diet, genetics and epigenetic factors may also have a role in the occurrence of the disease. Why H. pylori cause GC in only a minority of those infected remains unknown. Although mechanisms of H. pylori-induced carcinogenesis are not yet well understood, several genotypes of H. pylori have been associated with strain virulence and disease risk. Primary prevention of GC should be addressed by avoiding exposure to factors that increase the risk and to promote factors associated with decrease risk. Vaccines against H. pylori are an ongoing promise and not yet available. Chemoprevention through vitamin supplementation has shown no benefit. Screening and eradication of H. pylori in the general population is not advised. Given that GC is a multiple-steps process, the identification of patients with preneoplastic lesions with high risk of progression, and periodic endoscopic surveillance of them represents the most effective way for early diagnosis of GC. However, clinical guidelines for surveillance are lacking and there are no clear criteria to classify patients into high or low risk of progressing to GC. No study has shown the potential usefulness of combining the information on the type of preneoplastic lesions, genetic and epigenetic, lifestyle and virulence bacterial factors in order to identify high risk patients who need more intensive surveillance. The integration of all this information, in a prediction model requires further research and could be the most important contribution for reducing the burden of GC.
Collapse
Affiliation(s)
- Carlos A González
- Unit of Nutrition, Environment and Cancer, Epidemiology Research Programme, Catalan Institut of Oncology, Hospitalet del Llobregat, Barcelona, Spain.
| | | |
Collapse
|
113
|
Bilbo SD, Wray GA, Perkins SE, Parker W. Reconstitution of the human biome as the most reasonable solution for epidemics of allergic and autoimmune diseases. Med Hypotheses 2011; 77:494-504. [DOI: 10.1016/j.mehy.2011.06.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/04/2011] [Indexed: 02/07/2023]
|
114
|
The protective effect of the recombinant 53-kDa protein of Trichinella spiralis on experimental colitis in mice. Dig Dis Sci 2011; 56:2810-7. [PMID: 21476030 DOI: 10.1007/s10620-011-1689-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 03/21/2011] [Indexed: 12/26/2022]
Abstract
BACKGROUND Helminth infection has been proven to reduce the severity of experimental inflammatory bowel disease (IBD). The excretory-secretory proteins of helminths play an important role in the process of immunomodulation. AIMS In the present study, we aimed to investigate the protective potential of recombinant Trichinella spiralis (TS) 53-kDa protein (rTsP53), a component of excretory-secretory proteins, on experimental colitis in mice. METHODS BALB/c mice were treated subcutaneously with 50 μg rTsP53 three times at an interval of 5 days. Colitis was induced by intrarectal administration of 5 mg trinitrobenzene sulfonic acid (TNBS). Disease activities and macroscopic and microscopic scores were evaluated. To determine immune response provoked by rTsP53, we measured specific IgG1 and IgG2a values against rTsP53 in sera of mice. We also detected cytokine profiles as well as the markers of alternatively activated macrophages (M2) in mice. RESULTS RTsP53 ameliorated significantly the disease activity index (DAI) as well as the macroscopic and microscopic scores. IgG1 but not IgG2a was the predominant specific antibody detected in the sera of immunized mice, indicating the potential of stimulating T-helper (Th) 2 bias response by rTsP3. Pre-treatment with rTsP53 decreased serum Th1 cytokines (TNF-a, IFN-γ) and elevated serum levels of serum Th2 cytokines (IL-4, IL-13); it also decreased colonic Th1 cytokines (TNF-α, IL-6) and colonic regulatory cytokines (IL-10, TGF-β1). RTsP53 increased colonic M2 markers, arginase-1 (Arg-1), and found in inflammatory zone 1 (FIZZ1), compared to mice without rTsP53 pretreatment. CONCLUSIONS RTsP53 is a potential protective agent for IBD.
Collapse
|
115
|
Coinfection with Enterohepatic Helicobacter species can ameliorate or promote Helicobacter pylori-induced gastric pathology in C57BL/6 mice. Infect Immun 2011; 79:3861-71. [PMID: 21788386 DOI: 10.1128/iai.05357-11] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To investigate how different enterohepatic Helicobacter species (EHS) influence Helicobacter pylori gastric pathology, C57BL/6 mice were infected with Helicobacter hepaticus or Helicobacter muridarum, followed by H. pylori infection 2 weeks later. Compared to H. pylori-infected mice, mice infected with H. muridarum and H. pylori (HmHp mice) developed significantly lower histopathologic activity index (HAI) scores (P < 0.0001) at 6 and 11 months postinoculation (MPI). However, mice infected with H. hepaticus and H. pylori (HhHp mice) developed more severe gastric pathology at 6 MPI (P = 0.01), with a HAI at 11 MPI (P = 0.8) similar to that of H. pylori-infected mice. H. muridarum-mediated attenuation of gastritis in coinfected mice was associated with significant downregulation of proinflammatory Th1 (interlukin-1beta [Il-1β], gamma interferon [Ifn-γ], and tumor necrosis factor-alpha [Tnf-α]) cytokines at both time points and Th17 (Il-17A) cytokine mRNA levels at 6 MPI in murine stomachs compared to those of H. pylori-infected mice (P < 0.01). Coinfection with H. hepaticus also suppressed H. pylori-induced elevation of gastric Th1 cytokines Ifn-γ and Tnf-α (P < 0.0001) but increased Th17 cytokine mRNA levels (P = 0.028) at 6 MPI. Furthermore, mRNA levels of Il-17A were positively correlated with the severity of helicobacter-induced gastric pathology (HhHp>H. pylori>HmHp) (at 6 MPI, r² = 0.92, P < 0.0001; at 11 MPI, r² = 0.82, P < 0.002). Despite disparate effects on gastritis, colonization levels of gastric H. pylori were increased in HhHp mice (at 6 MPI) and HmHp mice (at both time points) compared to those in mono-H. pylori-infected mice. These data suggest that despite consistent downregulation of Th1 responses, EHS coinfection either attenuated or promoted the severity of H. pylori-induced gastric pathology in C57BL/6 mice. This modulation was related to the variable effects of EHS on gastric interleukin 17 (IL-17) responses to H. pylori infection.
Collapse
|
116
|
The Human Gastric Pathogen Helicobacter pylori and Its Association with Gastric Cancer and Ulcer Disease. ACTA ACUST UNITED AC 2011. [DOI: 10.1155/2011/340157] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
With the momentous discovery in the 1980's that a bacterium, Helicobacter pylori, can cause peptic ulcer disease and gastric cancer, antibiotic therapies and prophylactic measures have been successful, only in part, in reducing the global burden of these diseases. To date, ~700,000 deaths worldwide are still attributable annually to gastric cancer alone. Here, we review H. pylori's contribution to the epidemiology and histopathology of both gastric cancer and peptic ulcer disease. Furthermore, we examine the host-pathogen relationship and H. pylori biology in context of these diseases, focusing on strain differences, virulence factors (CagA and VacA), immune activation and the challenges posed by resistance to existing therapies. We consider also the important role of host-genetic variants, for example, in inflammatory response genes, in determining infection outcome and the role of H. pylori in other pathologies—some accepted, for example, MALT lymphoma, and others more controversial, for example, idiopathic thrombocytic purpura. More recently, intriguing suggestions that H. pylori has protective effects in GERD and autoimmune diseases, such as asthma, have gained momentum. Therefore, we consider the basis for these suggestions and discuss the potential impact for future therapeutic rationales.
Collapse
|
117
|
Ohtani M, Ge Z, García A, Rogers AB, Muthupalani S, Taylor NS, Xu S, Watanabe K, Feng Y, Marini RP, Whary MT, Wang TC, Fox JG. 17 β-estradiol suppresses Helicobacter pylori-induced gastric pathology in male hypergastrinemic INS-GAS mice. Carcinogenesis 2011; 32:1244-50. [PMID: 21565825 DOI: 10.1093/carcin/bgr072] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori-associated gastric cancer is male predominant and animal studies suggest that sex hormones influence gastric carcinogenesis. We investigated the effects of 17β-estradiol (E2) or castration on H.pylori-induced gastritis in male INS-GAS/FVB/N (Tg(Ins1-GAS)1Sbr) mice. Comparisons were made to previously evaluated sham (n = 8) and H.pylori-infected (n = 8), intact male INS-GAS mice which had developed severe corpus gastritis accompanied by atrophy, hyperplasia, intestinal metaplasia and dysplasia of the epithelium within 16 weeks postinfection (all P < 0.01). Castration at 8 weeks of age had no sparing effect on lesions in uninfected (n = 5) or H.pylori-infected mice (n = 7) but all lesion subfeatures were attenuated by E2 in H.pylori-infected mice (n = 7) (P < 0.001). Notably, inflammation was not reduced but glandular atrophy, hyperplasia, intestinal metaplasia and dysplasia were also less severe in uninfected, E2-treated mice (n = 7) (P < 0.01). Attenuation of gastric lesions by E2 was associated with lower messenger RNA (mRNA) expression of interferon (IFN)-γ (P < 0.05) and interleukin (IL)-1β (P < 0.004), and higher IL-10 (P < 0.02) as well as decreased numbers of Foxp3(+) regulatory T cells when compared with infected intact males. Infected E2-treated mice also developed higher Th2-associated anti-H.pylori IgG1 responses (P < 0.05) and significantly lower Ki-67 indices of epithelial proliferation (P < 0.05). E2 elevated expression of mRNA for Foxp3 (P < 0.0001) and IL-10 (P < 0.01), and decreased IL-1β (P < 0.01) in uninfected, intact male mice compared with controls. Therefore, estrogen supplementation, but not castration, attenuated gastric lesions in H.pylori-infected male INS-GAS mice and to a lesser extent in uninfected mice, potentially by enhancing IL-10 function, which in turn decreased IFN-γ and IL-1β responses induced by H.pylori.
Collapse
Affiliation(s)
- Masahiro Ohtani
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Perry S, Hussain R, Parsonnet J. The impact of mucosal infections on acquisition and progression of tuberculosis. Mucosal Immunol 2011; 4:246-51. [PMID: 21412228 PMCID: PMC5480373 DOI: 10.1038/mi.2011.11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
More than one-third of the world's population, or over 2 billion people, are infected with Mycobacterium tuberculosis, the causative pathogen of tuberculosis in humans. Why only 10% of those infected develop active disease while the remainder harbor latent infection remains one of the greatest scientific and public health mysteries. Bacterial persistence is characterized by a dynamic state of immunological tolerance between pathogen and host. The critical role of CD4(+) T cells in defense against intracellular pathogens became evident during epidemiological studies of HIV-1 infection, which showed a clear inverse relationship between CD4(+) T-cell count in peripheral blood and increased risk of infection with M. tuberculosis, pneumocystis and Toxoplasma gondii. There is also growing evidence of a common mucosal immune system, whereby immune cells activated at one mucosal site may disseminate to remote effector sites. In this commentary, we review emerging evidence from human studies that the outcome of M. tuberculosis infection is influenced by concurrent mucosal infections, using Helicobacter pylori and geohelminths as examples. Understanding how the complexity of microbial exposures influences host immunity may have important implications for vaccine development and therapeutic interventions.
Collapse
Affiliation(s)
- S Perry
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| | - R Hussain
- Department of Molecular Biology, Aga Khan University, Karachi, Pakistan
| | - J Parsonnet
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
119
|
Duodenal helminth infection alters barrier function of the colonic epithelium via adaptive immune activation. Infect Immun 2011; 79:2285-94. [PMID: 21444669 DOI: 10.1128/iai.01123-10] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic infection with intestinal helminth parasites is a major public health problem, particularly in the developing world, and can have significant effects on host physiology and the immune response to other enteric infections and antigens. The mechanisms underlying these effects are not well understood. In the current study, we investigated the impact of infection with the murine nematode parasite Heligmosomoides polygyrus, which resides in the duodenum, on epithelial barrier function in the colon. We found that H. polygyrus infection produced a significant increase in colonic epithelial permeability, as evidenced by detection of elevated serum levels of the tracer horseradish peroxidase following rectal administration. This loss of normal barrier function was associated with clear ultrastructural changes in the tight junctions of colonic epithelial cells and an alteration in the expression and distribution of the junctional protein E-cadherin. These parasite-induced abnormalities were not observed in SCID mice but did occur in SCID mice that were adoptively transferred with wild-type T cells, indicating a requirement for adaptive immunity. Furthermore, the helminth-induced increase in gut permeability was not seen in STAT6 knockout (KO) mice. Taken together, the results demonstrate that one of the mechanisms by which helminths exert their effects involves the lymphocyte- and STAT6-dependent breakdown of the intestinal epithelial barrier. This increase in epithelial permeability may facilitate the movement of lumenal contents across the mucosa, thus helping to explain how helminth infection can alter the immune response to enteric antigens.
Collapse
|
120
|
Arnold IC, Zigova Z, Holden M, Lawley TD, Rad R, Dougan G, Falkow S, Bentley SD, Müller A. Comparative whole genome sequence analysis of the carcinogenic bacterial model pathogen Helicobacter felis. Genome Biol Evol 2011; 3:302-8. [PMID: 21402865 PMCID: PMC4197744 DOI: 10.1093/gbe/evr022] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The gram-negative bacterium Helicobacter felis naturally
colonizes the gastric mucosa of dogs and cats. Due to its ability to
persistently infect laboratory mice, H. felis has been used
extensively to experimentally model gastric disorders induced in humans by
H. pylori. We determined the 1.67 Mb genome sequence of
H. felis using combined Solexa and 454 pyrosequencing,
annotated the genome, and compared it with multiple previously published
Helicobacter genomes. About 1,063 (63.6%) of the 1,671
genes identified in the H. felis genome have orthologues in
H. pylori, its closest relative among the fully sequenced
Helicobacter species. Many H. pylori
virulence factors are shared by H. felis: these include the
gamma-glutamyl transpeptidase GGT, the immunomodulator NapA, and the secreted
enzymes collagenase and HtrA. Helicobacter felis lacks a Cag
pathogenicity island and the vacuolating cytotoxin VacA but possesses a complete
comB system conferring natural competence. Remarkable features of the H.
felis genome include its paucity of transcriptional regulators and
an extraordinary abundance of chemotaxis sensors and restriction/modification
systems. Helicobacter felis possesses an episomally replicating
6.7-kb plasmid and harbors three chromosomal regions with deviating GC content.
These putative horizontally acquired regions show homology and synteny with the
recently isolated H. pylori plasmid pHPPC4 and homology to
Campylobacter bacteriophage genes (transposases,
structural, and lytic genes), respectively. In summary, the H.
felis genome harbors a variety of putative mobile elements that are
unique among Helicobacter species and may contribute to this
pathogen’s carcinogenic properties.
Collapse
Affiliation(s)
- Isabelle C Arnold
- Institute of Molecular Cancer Research, University of Zürich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
121
|
|
122
|
Blackwell AD, Snodgrass JJ, Madimenos FC, Sugiyama LS. Life history, immune function, and intestinal helminths: Trade-offs among immunoglobulin E, C-reactive protein, and growth in an Amazonian population. Am J Hum Biol 2011; 22:836-48. [PMID: 20865759 DOI: 10.1002/ajhb.21092] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVES Infection with helminths is associated with shifts in host immunity, including increased production of immunoglobulin E (IgE) and reduced inflammation. Given limited energy budgets, these shifts may involve changes in energy allocation toward competing demands. Here we test for potential trade-offs between growth, IgE, and the inflammatory marker C-reactive protein (CRP). METHODS Dried blood spots and anthropometrics were collected from 162 Shuar forager-horticulturalists from a village in southeastern Ecuador. Enzyme-linked immunosorbent assays (ELISAs) were used to measure IgE and CRP. Relationships among IgE, CRP, and anthropometrics were examined in three groups: children aged 2-7 years (n = 63), children aged 8-15 (n = 61), and adults over age 18 (n = 37). RESULTS Geometric mean IgE was 1,196 IU ml⁻¹ while geometric mean CRP was 1.33 mg l⁻¹. In children, IgE and CRP were negatively correlated (r = -0.21, P = 0.02, df = 122). Controlling for fat stores and age, IgE was associated with lower stature in children (t = -2.04, P = 0.04, df = 109), and adults (t = -3.29, P < 0.01, df = 33). In children there was a significant interaction between age and CRP, such that in younger children CRP was associated with shorter stature, but in older children was associated with greater stature (t = 2.15, P = 0.04, df = 109). CONCLUSIONS These results suggest that infection with helminths may have hidden costs associated with immunological changes, and that these costs may ultimately affect growth and other life history parameters.
Collapse
Affiliation(s)
- Aaron D Blackwell
- Integrative Anthropological Sciences, University of California, Santa Barbara, California 93106-3210, USA.
| | | | | | | |
Collapse
|
123
|
|
124
|
Lofgren JL, Whary MT, Ge Z, Muthupalani S, Taylor NS, Mobley M, Potter A, Varro A, Eibach D, Suerbaum S, Wang TC, Fox JG. Lack of commensal flora in Helicobacter pylori-infected INS-GAS mice reduces gastritis and delays intraepithelial neoplasia. Gastroenterology 2011; 140:210-20. [PMID: 20950613 PMCID: PMC3006487 DOI: 10.1053/j.gastro.2010.09.048] [Citation(s) in RCA: 272] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 08/31/2010] [Accepted: 09/17/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Transgenic FVB/N insulin-gastrin (INS-GAS) mice have high circulating gastrin levels, and develop spontaneous atrophic gastritis and gastrointestinal intraepithelial neoplasia (GIN) with 80% prevalence 6 months after Helicobacter pylori infection. GIN is associated with gastric atrophy and achlorhydria, predisposing mice to nonhelicobacter microbiota overgrowth. We determined if germfree INS-GAS mice spontaneously develop GIN and if H pylori accelerates GIN in gnotobiotic INS-GAS mice. METHODS We compared gastric lesions, levels of messenger RNA, serum inflammatory mediators, antibodies, and gastrin among germfree and H pylori-monoinfected INS-GAS mice. Microbiota composition of specific pathogen-free (SPF) INS-GAS mice was quantified by pyrosequencing. RESULTS Germfree INS-GAS mice had mild hypergastrinemia but did not develop significant gastric lesions until 9 months old and did not develop GIN through 13 months. H pylori monoassociation caused progressive gastritis, epithelial defects, oxyntic atrophy, marked foveolar hyperplasia, dysplasia, and robust serum and tissue proinflammatory immune responses (particularly males) between 5 and 11 months postinfection (P<0.05, compared with germfree controls). Only 2 of 26 female, whereas 8 of 18 male, H pylori-infected INS-GAS mice developed low to high-grade GIN by 11 months postinfection. Stomachs of H pylori-infected SPF male mice had significant reductions in Bacteroidetes and significant increases in Firmicutes. CONCLUSIONS Gastric lesions take 13 months longer to develop in germfree INS-GAS mice than male SPF INS-GAS mice. H pylori monoassociation accelerated gastritis and GIN but caused less severe gastric lesions and delayed onset of GIN compared with H pylori-infected INS-GAS mice with complex gastric microbiota. Changes in gastric microbiota composition might promote GIN in achlorhydric stomachs of SPF mice.
Collapse
Affiliation(s)
- Jennifer L. Lofgren
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Mark T. Whary
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Zhongming Ge
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Sureshkumar Muthupalani
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Nancy S. Taylor
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Melissa Mobley
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Amanda Potter
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Andrea Varro
- Physiological Laboratory, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Daniel Eibach
- Medizinische Hochschule Hannover, Institut für Medizinische Mikrobiologie und Krankenhaushygiene, D-30625 Hannover, Germany
| | - Sebastian Suerbaum
- Medizinische Hochschule Hannover, Institut für Medizinische Mikrobiologie und Krankenhaushygiene, D-30625 Hannover, Germany
| | - Timothy C. Wang
- Medizinische Hochschule Hannover, Institut für Medizinische Mikrobiologie und Krankenhaushygiene, D-30625 Hannover, Germany
| | - James G. Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
125
|
Helicobacter pylori and gastric cancer: factors that modulate disease risk. Clin Microbiol Rev 2010; 23:713-39. [PMID: 20930071 DOI: 10.1128/cmr.00011-10] [Citation(s) in RCA: 964] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori is a gastric pathogen that colonizes approximately 50% of the world's population. Infection with H. pylori causes chronic inflammation and significantly increases the risk of developing duodenal and gastric ulcer disease and gastric cancer. Infection with H. pylori is the strongest known risk factor for gastric cancer, which is the second leading cause of cancer-related deaths worldwide. Once H. pylori colonizes the gastric environment, it persists for the lifetime of the host, suggesting that the host immune response is ineffective in clearing this bacterium. In this review, we discuss the host immune response and examine other host factors that increase the pathogenic potential of this bacterium, including host polymorphisms, alterations to the apical-junctional complex, and the effects of environmental factors. In addition to host effects and responses, H. pylori strains are genetically diverse. We discuss the main virulence determinants in H. pylori strains and the correlation between these and the diverse clinical outcomes following H. pylori infection. Since H. pylori inhibits the gastric epithelium of half of the world, it is crucial that we continue to gain understanding of host and microbial factors that increase the risk of developing more severe clinical outcomes.
Collapse
|
126
|
Zavros Y, Mesiwala N, Waghray M, Todisco A, Shulkes A, Merchant JL. Histamine 3 receptor activation mediates inhibition of acid secretion during Helicobacter-induced gastritis. World J Gastrointest Pathophysiol 2010; 1:154-65. [PMID: 21607157 PMCID: PMC3097961 DOI: 10.4291/wjgp.v1.i5.154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 11/24/2010] [Accepted: 12/01/2010] [Indexed: 02/07/2023] Open
Abstract
AIM: To test the hypothesis that histamine 3 receptor (H3R) activation during Helicobacter infection inhibits gastric acid secretion in vivo and in vitro.
METHODS: Helicobacter felis (H. felis) infected and uninfected C57Bl/6 mice were infused with either PBS or the H3 receptor antagonist thioperamide (THIO) for 12 wk. After treatment, mice were analyzed for morphological changes and gastric acid content. Total RNA was prepared from the stomachs of each group and analyzed for changes in somatostatin and gastrin mRNA abundance by real time-polymerase chain reaction (RT-PCR). Location of H3 receptors in the stomach was analyzed by co-localization using antibodies specific for the H3 receptor and parietal cell marker H+, K+-ATPase β subunit.
RESULTS: Inflammation and parietal cell atrophy was observed after 12 wk of H. felis infection. Interestingly, treatment with the H3R antagonist thioperamide (THIO) prior to and during infection prevented H. felis-induced inflammation and atrophy. Compared to the uninfected controls, infected mice also had significantly decreased gastric acid. After eradication of H. felis with THIO treatment, gastric acidity was restored. Compared to the control mice, somatostatin mRNA abundance was decreased while gastrin gene expression was elevated during infection. Despite elevated gastric acid levels, after eradication of H. felis with THIO, somatostatin mRNA was elevated whereas gastrin mRNA was suppressed. Immunofluorescence revealed the presence of H3 receptors on the parietal cells, somatostatin-secreting D-cells as well as the inflammatory cells.
CONCLUSION: This study shows that during H. felis infection, gastric acidity is suppressed as a consequence of an inhibitory effect on the parietal cell by H3R activation. The stimulation of gastric mucosal H3Rs increases gastrin expression and release by inhibiting release of somatostatin.
Collapse
|
127
|
Abstract
The role of infectious agents and chronic inflammation in carcinogenesis is being increasingly recognized. It has been estimated that about 18% of cancers are directly linked to infections, particularly gastric adenocarcinoma (Helicobacter pylori), cervical carcinoma (human papilloma viruses), and hepatocarcinoma (hepatitis B and C viruses). Multiple clinical trials of COX-2 inhibitors and other antiinflammatory agents have shown a beneficial effect on the development of diverse tumors, such as those of the colon, stomach, prostate, and breast. However, their mechanism of action is not completely understood and may differ among the infectious agents and tumor types. Because gastric adenocarcinomas account for more than 90% of all gastric malignancies, this review focuses on adenocarcinomas.
Collapse
Affiliation(s)
- M Blanca Piazuelo
- Division of Gastroenterology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | | | | |
Collapse
|
128
|
Qadura M, Waters B, Burnett E, Chegeni R, Hough C, Othman M, Lillicrap D. Immunoglobulin isotypes and functional anti-FVIII antibodies in response to FVIII treatment in Balb/c and C57BL/6 haemophilia A mice. Haemophilia 2010; 17:288-95. [PMID: 21091850 DOI: 10.1111/j.1365-2516.2010.02397.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Previous studies have demonstrated that genetic factors play an important role in determining the likelihood of formation of anti-factor VIII (FVIII) antibodies in haemophilia A patients. We were interested in characterizing the spectrum of FVIII antibody formation and the primary and secondary immune responses after FVIII administration in two different exon 16-disrupted haemophilia A mouse strains, Balb/c and C57BL/6. Balb/c and C57BL/6 E16 haemophilia A mice were used in all experiments. Total FVIII antibodies and FVIII inhibitors were measured using ELISA and Bethesda assays respectively. T- and B-cell cytokines were quantified using ELISA and flow cytometry. FVIII antibodies, but not functional inhibitors were detectable 1 week after the first FVIII treatment in both strains. These antibodies mainly belonged to the IgM and IgA isotypes. After the fourth FVIII treatment, neutralizing anti-FVIII antibodies were detected in both mouse strains: Balb/c (mean inhibitory titer 58 BU) and C57BL/6 (mean inhibitory titer 82 BU). IgG1 levels were similar in both strains but the IgG2A and IgG2B subclasses were higher in C57BL/6 mice. The results of intracellular cytokine staining of T cells indicated that the FVIII-treated C57BL/6 mice produced more IL10 and Th1 cytokines than the FVIII-treated Balb/c mice. These studies show that C57BL/6 mice develop a stronger immune response towards FVIII than Balb/c mice. We propose that the enhanced Th1 and IL10 cytokine micro-environment induced in C57BL/6 mice is responsible for this difference. Therefore, genetic strain-dependent differences must be considered when evaluating immunological outcomes in mouse models of haemophilia A.
Collapse
Affiliation(s)
- M Qadura
- Department of Pathology and Molecular Medicine, Richardson Laboratory, Queen's University, Kingston, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
129
|
Amedei A, Codolo G, Del Prete G, de Bernard M, D'Elios MM. The effect of Helicobacter pylori on asthma and allergy. J Asthma Allergy 2010; 3:139-47. [PMID: 21437048 PMCID: PMC3047919 DOI: 10.2147/jaa.s8971] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Indexed: 12/12/2022] Open
Abstract
Current evidence indicates an inverse association between Helicobacter pylori and asthma and allergy. H. pylori is a Gram-negative bacterium which represents the major cause of peptic ulcer and gastric cancer, and preferentially elicits a T helper (Th)-1 response. Many H. pylori factors, such as the neutrophil-activating factor of H. pylori (HP-NAP), are able to drive Th-1 polarization and to display a powerful inhibition of allergic Th-2 response. This article proposes an overview of the actual knowledge about the effects of H. pylori on asthma and allergy. Special attention has been drawn to HP-NAP as a potential novel strategy for the prevention and treatment of asthma and atopy.
Collapse
Affiliation(s)
- Amedeo Amedei
- Policlinico AOU Careggi, Department Internal Medicine, University of Florence, Italy
| | | | | | | | | |
Collapse
|
130
|
Perez-Perez GI, Maw AM, Feingold-Link L, Gunn J, Bowers AL, Minano C, Rautelin H, Kosunen TU, Blaser MJ. Longitudinal analysis of serological responses of adults to Helicobacter pylori antigens. J Infect Dis 2010; 202:916-23. [PMID: 20698790 PMCID: PMC2924458 DOI: 10.1086/655660] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Because Helicobacter pylori persist for decades in the human stomach, the aim of this study was to examine the long-term course of H. pylori-specific serum immunoglobulin G (IgG) responses with respect to subclass and antigenic target. We studied paired serum samples obtained in 1973 and in 1994 in Vammala, Finland, from 64 healthy H. pylori-positive adults and from other healthy control subjects. H. pylori serum immunoglobulin A, IgG, and IgG subclass responses were determined by antigen-specific enzyme-linked immunosorbent assays. H. pylori-specific IgG1 and IgG4 subtype responses from 47 subjects were similar in 1973 and 1994, but not when compared with unrelated persons. H. pylori-specific IgG1:IgG4 ratios among the participants varied >1000-fold; however, 57 (89.1%) of 64 subjects had an IgG1:IgG4 ratio >1.0, consistent with a predominant IgG1 (Th1) response. Furthermore, ratios in individual hosts were stable over the 21-year period (r = 0.56; P < .001). The immune response to heat shock protein HspA was unchanged in 49 (77%) of the 64 subjects tested; of the 15 whose serostatus changed, all seroconverted and were significantly younger than those whose status did not change. These findings indicate that H. pylori-specific antibody responses are host-specific with IgG1:IgG4 ratios stable over 21 years, IgG1 responses predominating, and HspA seroconversion with aging.
Collapse
Affiliation(s)
- Guillermo I Perez-Perez
- Department of Medicine, New York University Langone Medical Center, New York, New York, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Toller IM, Altmeyer M, Kohler E, Hottiger MO, Müller A. Inhibition of ADP ribosylation prevents and cures helicobacter-induced gastric preneoplasia. Cancer Res 2010; 70:5912-22. [PMID: 20634404 DOI: 10.1158/0008-5472.can-10-0528] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gastric adenocarcinoma develops as a consequence of chronic inflammation of the stomach lining that is caused by persistent infection with the bacterium Helicobacter pylori. Gastric carcinogenesis progresses through a sequence of preneoplastic lesions that manifest histologically as atrophic gastritis, intestinal metaplasia, and dysplasia. We show here in several preclinical models of Helicobacter-induced atrophic gastritis, epithelial hyperplasia, and metaplasia that the inhibition of ADP ribosylation by the small-molecule inhibitor PJ34 not only prevents the formation of gastric cancer precursor lesions, but also efficiently reverses preexisting lesions. PJ34 exerts its chemopreventive and therapeutic effects by impairing Helicobacter-specific T-cell priming and T(H)1 polarization in the gut-draining mesenteric lymph nodes. The subsequent infiltration of pathogenic T cells into the gastric mucosa and the ensuing gastric T cell-driven immunopathology are prevented efficiently by PJ34. Our data indicate that PJ34 directly suppresses T-cell effector functions by blocking the IFN-gamma production of mesenteric lymph node T cells ex vivo. Upon exposure to PJ34, purified T cells failed to synthesize ADP-ribose polymers and to activate the transcription of genes encoding IFN-gamma, interleukin 2, and the interleukin 2 receptor alpha chain in response to stimuli such as CD3/CD28 cross-linking or phorbol 12-myristate 13-acetate/ionomycin. The immunosuppressive and chemoprotective effects of PJ34 therefore result from impaired T-cell activation and T(H)1 polarization, and lead to the protection from preneoplastic gastric immunopathology. In conclusion, ADP-ribosylating enzymes constitute novel targets for the treatment of Helicobacter-associated gastric lesions predisposing infected individuals to gastric cancer and may also hold promise for the treatment of other T cell-driven chronic inflammatory conditions and autoimmune pathologies.
Collapse
Affiliation(s)
- Isabella M Toller
- Institute of Molecular Cancer Research and Institute of Veterinary Biochemistry and Molecular Biology, University of Zürich, Zürich, Switzerland
| | | | | | | | | |
Collapse
|
132
|
Mutagenic potency of Helicobacter pylori in the gastric mucosa of mice is determined by sex and duration of infection. Proc Natl Acad Sci U S A 2010; 107:15217-22. [PMID: 20699385 DOI: 10.1073/pnas.1009017107] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Helicobacter pylori is a human carcinogen, but the mechanisms evoked in carcinogenesis during this chronic inflammatory disease remain incompletely characterized. We determined whether chronic H. pylori infection induced mutations in the gastric mucosa of male and female gpt delta C57BL/6 mice infected for 6 or 12 mo. Point mutations were increased in females infected for 12 mo. The mutation frequency in this group was 1.6-fold higher than in uninfected mice of both sexes (P < 0.05). A:T-to-G:C transitions and G:C-to-T:A transversions were 3.8 and 2.0 times, respectively, more frequent in this group than in controls. Both mutations are consistent with DNA damage induced by oxidative stress. No increase in the frequency of deletions was observed. Females had more severe gastric lesions than males at 6 mo postinfection (MPI; P < 0.05), but this difference was absent at 12 MPI. In all mice, infection significantly increased expression of IFNgamma, IL-17, TNFalpha, and iNOS at 6 and 12 mo, as well as H. pylori-specific IgG1 levels at 12 MPI (P < 0.05) and IgG2c levels at 6 and 12 MPI (P < 0.01 and P < 0.001). At 12 MPI, IgG2c levels in infected females were higher than at 6 MPI (P < 0.05) and also than those in infected males at 12 MPI (P < 0.05). Intensity of responses was mediated by sex and duration of infection. Lower H. pylori colonization indicated a more robust host response in females than in males. Earlier onset of severe gastric lesions and proinflammatory, Th1-biased responses in female C57BL/6 mice may have promoted mutagenesis by exposing the stomach to prolonged oxidative stress.
Collapse
|
133
|
Cheng HH, Chang CS, Wang HJ, Wang WC. Interleukin-1beta and -10 polymorphisms influence erosive reflux esophagitis and gastritis in Taiwanese patients. J Gastroenterol Hepatol 2010; 25:1443-51. [PMID: 20659236 DOI: 10.1111/j.1440-1746.2010.06310.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Helicobacter pylori (H. pylori) infection induces cytokine production and is associated with gastrointestinal diseases. This study examined the relationship of gene polymorphisms, including interleukin (IL)-1beta, -10, -8, and tumor necrosis factor-alpha (TNF-alpha), H. pylori infection, and susceptibility to gastrointestinal disorders in Taiwanese patients. METHODS IL-1beta-511/-31/+3953, -10-1082/-819/-592, -8-251, and TNF-alpha-308 polymorphisms were assessed in 628 gastrointestinal disease patients, and 176 healthy controls were analyzed using the polymerase chain reaction-restriction fragment length polymorphism method. RESULTS IL-1beta-511 T/T and -31 C/C genotypes, and IL-1beta-511 T and -31 C alleles were associated with an increased risk of reflux esophagitis (P = 0.034, odds ratio [OR] = 1.384, 95% confidence interval [CI]: 1.023-1.871; P = 0.031, OR = 1.388, 95% CI: 1.028-1.873; P = 0.044, OR = 1.342, 95% CI: 1.008-1.786; and P = 0.040, OR = 1.349, 95% CI: 1.014-1.796, respectively). No relationship was found between H. pylori infection and the risk of reflux esophagitis. IL-10-819 C/T and -10-592 A/C genotypes and IL-10-1082/-819/-592 ATA/ACC and ATA/GCC haplotypes were associated with an increased risk of gastritis (P = 0.021, OR = 1.721, 95% CI: 1.084-2.733; P = 0.016, OR = 1.766, 95% CI: 1.112-2.805; P = 0.039, OR = 1.662, 95% CI: 1.024-2.697; and P = 0.035, OR = 1.600, 95% CI: 1.024-2.499, respectively). CONCLUSION Among Taiwanese patients, IL-1beta and -10 polymorphisms were associated with an increased risk of erosive reflux esophagitis and gastritis, respectively.
Collapse
Affiliation(s)
- Hsin-Hung Cheng
- Institute of Molecular and Cellular Biology and Department of Life Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | | | | | | |
Collapse
|
134
|
Rausch S, Held J, Stange J, Lendner M, Hepworth MR, Klotz C, Lucius R, Pogonka T, Hartmann S. A matter of timing: Early, not chronic phase intestinal nematode infection restrains control of a concurrent enteric protozoan infection. Eur J Immunol 2010; 40:2804-15. [DOI: 10.1002/eji.201040306] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
135
|
Abstract
Chronic inflammation is essential for cancer growth and metastasis. It follows that factors reducing inflammation would abrogate cancer and restore tissue health. However, roles for anti-inflammatory CD4+ regulatory cells (T(REG)) in cancer are enigmatic and controversial. Our recent data reveal that T(REG) may function in cancer similarly to inflammatory bowel disease or multiple sclerosis, whereby T(REG) accumulate but lack potency to restore tissue homeostasis under inflammatory conditions. Interestingly, early life exposures to diverse environmental organisms reinforce a protective T(REG) phenotype that inhibits cancer. In contrast, hygienic individuals with few exposures earlier in life suffer from a dysregulated T(REG) feedback loop. Consequently, hygienic subjects have increased risk of malignancy later in life. This cancer condition is reversible by blocking underlying inflammation. Taken together, these data help explain increased inflammation-associated cancer rates in hygienic societies and identify targets to abrogate cancer and restore overall health.
Collapse
Affiliation(s)
- Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | |
Collapse
|
136
|
Cytotoxic T cells in H. pylori-related gastric autoimmunity and gastric lymphoma. J Biomed Biotechnol 2010; 2010:104918. [PMID: 20617132 PMCID: PMC2896618 DOI: 10.1155/2010/104918] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 04/28/2010] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori infection is the major cause of gastroduodenal pathologies, but only a minority of infected patients develop gastric B-cell lymphoma, gastric autoimmunity, or other life threatening diseases, as gastric cancer or peptic ulcer. The type of host immune response against H. pylori, particularly the cytolytic effector functions of T cells, is crucial for the outcome of the infection. T cells are potentially able to kill a target via different mechanisms, such as perforins or Fas-Fas ligand interaction. In H. pylori-infected patients with gastric autoimmunity cytolytic T cells, that cross-recognize different epitopes of H. pylori proteins and H(+)K(+)-ATPase autoantigen, infiltrate the gastric mucosa and lead to gastric atrophy via long-lasting activation of Fas ligand-mediated appotosis and perforin-induced cytotoxicity. On the other hand, gastric T cells from MALT lymphoma exhibit defective perforin- and Fas-Fas ligand-mediated killing of B cells, with consequent abnormal help for B-cell proliferation, suggesting that deregulated and exhaustive H. pylori-induced T cell-dependent B-cell activation can support both the onset and the promotion of low-grade B-cell lymphoma.
Collapse
|
137
|
D'Elios MM, de Bernard M. To treat or not to treat Helicobacter pylori to benefit asthma patients. Expert Rev Respir Med 2010; 4:147-50. [PMID: 20406078 DOI: 10.1586/ers.10.9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
138
|
The enigma of Helicobacter pylori infection and gastric cancer. Indian J Gastroenterol 2010; 29:95-100. [PMID: 20585917 PMCID: PMC3217495 DOI: 10.1007/s12664-010-0024-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 06/07/2010] [Indexed: 02/04/2023]
Abstract
Although H. pylori has been recognized as a class I carcinogen, incongruence between infection prevalence and cancer incidence has been reported. Holcombe called attention to the high prevalence of infection in the face of low cancer rates, which he called "The African Enigma". Similar observations have now been made in other geographic areas. Gastric carcinoma should be considered an infectious disease, for which the classical epidemiologic model of causality applies. The model proposes that tissue injury inflicted by the infectious agent is modulated by its interactions with host and environmental factors. Although approximately half of the world's population is infected, only a small proportion of people develop cancer. The African enigma is a striking example of the major contrasts in cancer risk among populations with similarly high prevalence of infection. The mechanisms involved in reducing the risk of cancer in infected individuals are explored in this article, which may lead to the design of effective prevention strategies.
Collapse
|
139
|
Martin HR, Shakya KP, Muthupalani S, Ge Z, Klei TR, Whary MT, Fox JG. Brugia filariasis differentially modulates persistent Helicobacter pylori gastritis in the gerbil model. Microbes Infect 2010; 12:748-58. [PMID: 20685294 DOI: 10.1016/j.micinf.2010.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 04/21/2010] [Accepted: 05/10/2010] [Indexed: 01/15/2023]
Abstract
In select Helicobacter pylori-infected populations with low gastric cancer, nematode coinfections are common and both helicobacter gastritis and filariasis are modeled in gerbils. We evaluated gastritis, worm counts, tissue cytokine gene expression levels and Th1/Th2-associated antibody responses in H. pylori and Brugia pahangi mono- and coinfected gerbils. H. pylori-associated gastritis indices were significantly lower 21 weeks post-infection in coinfected gerbils (p < or = 0.05) and were inversely proportional to worm counts (r(2) = -0.62, p < 0.003). Additionally, IFN-gamma, IL-1 beta, CXCL1, IL-4 and IL-10 mRNA levels in the gastric antrum reflected a significant host response to gastric H. pylori and as well as systemic filariasis (p < or = 0.05). Despite increasing worm burden (p < 0.05), gastritis progressed in coinfected gerbils (p < 0.03) becoming equivalent to H. pylori-infected gerbils at 42 weeks (p = 0.7). Pro- and anti-inflammatory mediator mRNA levels were notably downregulated in B. pahangi infected gerbils below uninfected control values, suggesting hyporesponsiveness to B. pahangi. Consistent with an increasing Th1 response to H. pylori, IgG2a (p < 0.01), IL-1 beta (p = 0.04) and CXCL1 (p = 0.006) responses significantly increased and IL-4 (p = 0.05) and IL-10 (p = 0.04) were decreased in coinfected gerbils at 42 weeks. Initial systemic responses to B. pahangi resulted in attenuated gastritis in coinfected gerbils, but subsequent filarid-associated hyporesponsiveness appears to have promoted H. pylori gastritis.
Collapse
Affiliation(s)
- Heather R Martin
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | |
Collapse
|
140
|
Wu WKK, Cho CH, Lee CW, Fan D, Wu K, Yu J, Sung JJY. Dysregulation of cellular signaling in gastric cancer. Cancer Lett 2010; 295:144-53. [PMID: 20488613 DOI: 10.1016/j.canlet.2010.04.025] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 04/26/2010] [Accepted: 04/27/2010] [Indexed: 02/07/2023]
Abstract
The pathogenesis of gastric cancer is complex and related to multiple factors. Dysregulation of intracellular signaling pathways represents a common pathogenic mechanism and may be amenable to drug targeting. Multiple well-established oncogenic pathways, such as those mediated by cell cycle regulators, nuclear factor-kappaB, cyclooxygenase-2 and epidermal growth factor receptor are implicated in gastric carcinogenesis. Emerging evidence also underscores the importance of signaling pathways involved in the developmental process, including transforming growth factor-beta/bone morphogenetic protein signaling, Wnt/beta-catenin signaling, Hedgehog signaling and Notch signaling. Understanding their biological significance will provide a rational basis for drug development. Their relative importance and cross-talk in gastric carcinogenesis, however, are still not completely understood and warrant further investigation.
Collapse
Affiliation(s)
- William K K Wu
- LKS Institute of Health, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, China.
| | | | | | | | | | | | | |
Collapse
|
141
|
Belkaid Y, Liesenfeld O, Maizels RM. 99th Dahlem conference on infection, inflammation and chronic inflammatory disorders: induction and control of regulatory T cells in the gastrointestinal tract: consequences for local and peripheral immune responses. Clin Exp Immunol 2010; 160:35-41. [PMID: 20415849 DOI: 10.1111/j.1365-2249.2010.04131.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Regulatory T cells play a crucial role in normal gut homeostasis, as well as during infection with microbial or parasitic pathogens. Prior to infection, interactions with the commensal microflora are essential to differentiation of a healthy steady-state level of immunoregulation, mediated through both Toll-like receptor-dependent and -independent pathways. The ingress of pathogenic organisms may, according to the context, promote or reverse the regulatory environment, with onward consequences for inflammation in both the intestinal and extra-intestinal settings. Appropriate regulation of gut immunity thus depends upon a complex three-way interplay between host cells, commensals and pathogens, and can exert a major impact on systemic responses including allergy and autoimmunity.
Collapse
Affiliation(s)
- Y Belkaid
- NIAID, National Institutes of Health, Laboratory of Parasitic Diseases, Bethesda, MD, USA
| | | | | |
Collapse
|
142
|
Hamajima N, Hishida A. Genetic traits for the persistence of Helicobacter pylori infection. Per Med 2010; 7:249-262. [PMID: 29776221 DOI: 10.2217/pme.10.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori infection elevates the risk of gastric diseases, including peptic ulcer and gastric cancer. Persistent infection is the first step to induce H. pylori-induced multistage diseases. Although the roles of genetic traits on persistent infection have not yet been elucidated, some individuals escape from persistent infection. Possible favorable conditions for H. pylori seem to be low acid secretion, reduced innate immune responses, and easier binding to gastric epithelial cells. IL-1β and TNF-α inhibit acid secretion. The genetic polymorphisms associated with both molecules have the potential to be the genetic traits underlying persistent infection. Functional polymorphisms associated with innate immune responses could also be involved with the genetic traits, but no polymorphisms with consistent associations have been identified so far. The polymorphisms associated with molecules for adhesion to epithelial cells are candidates of genetic traits, but more research is needed.
Collapse
Affiliation(s)
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
143
|
Hosseini ME, Oghalaie A, Habibi G, Nahvijoo A, Hosseini ZM, Tashakoripoor M, Mohammadi M. Molecular detection of host cytokine expression in Helicobacter pylori infected patients via semi-quantitative RT-PCR. Indian J Med Microbiol 2010; 28:40-4. [PMID: 20061762 DOI: 10.4103/0255-0857.58727] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Helicobacter pylori (Hp) is a bacterium recognised as a main causative agent for the development of chronic active gastritis, peptic ulcer disease, gastric adenocarcinoma and primary gastric lymphoma. OBJECTIVE Determination of the levels of IFN-gamma (pro-inflammatory) and IL-4 (anti inflammatory) cytokine expression as indicators of Th1 and Th2 immune responses in gastric cancer (GC) and non gastric cancer (Non GC) dyspeptic patients by gene specific RT-PCR. MATERIALS AND METHODS Biopsy specimens were collected from three groups of gastric cancer (GC=18), non ulcer dyspepsia (NUD = 38) and peptic ulcer patients (PUD=20). Total RNA was extracted and complementary DNA was synthesised. PCR amplification was performed for HPRT, IFN-gamma and IL-4 cytokines and the intensity of each band was measured by densitometry and normalized against HPRT expression as a house keeping gene. RESULTS Comparison of the results from different groups of patients indicated that IFN-gamma gene expression was similar in nonGC dyspeptic patients (NUD and PUD groups; 3.38 +/- 0.57,3.43 +/- 0.41, respectively) whereas, in GC patients, it was significantly higher than others (5.52 +/- 0.59; P < 0.0001). On the other hand, IL-4 gene expression showed no significant difference between NUD and GC patients (2.81 +/- 0.43,2.3 +/- 0.12 respectively), whereas the expression rate of this cytokine was significantly higher in PUD patients (3.7 +/- 0.1; P 0.05). Our data indicate an association between Th1 and Th2 immune responses and the development of gastric cancer and peptic ulcer disease respectively.
Collapse
Affiliation(s)
- M Eshagh Hosseini
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran 13164, Iran
| | | | | | | | | | | | | |
Collapse
|
144
|
Noland GS, Chowdhury DR, Urban JF, Zavala F, Kumar N. Helminth infection impairs the immunogenicity of a Plasmodium falciparum DNA vaccine, but not irradiated sporozoites, in mice. Vaccine 2010; 28:2917-23. [PMID: 20188676 DOI: 10.1016/j.vaccine.2010.02.055] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 02/12/2010] [Accepted: 02/15/2010] [Indexed: 10/19/2022]
Abstract
Development of an effective vaccine against malaria remains a priority. However, a significant number of individuals living in tropical areas are also likely to be co-infected with helminths, which are known to adversely affect immune responses to a number of different existing vaccines. Here we compare the response to two prototype malaria vaccines: a transmission blocking DNA vaccine based on Pfs25, and a pre-erythrocytic malaria vaccine based on irradiated sporozoites in mice infected with the intestinal nematode Heligmosomoides polygyrus. Following primary immunization with Pfs25 DNA vaccine, levels of total IgG, as well as IgG1, IgG2a, IgG2b (all P=0.0002), and IgG3 (P=0.03) Pfs25 antibodies were significantly lower in H. polygyrus-infected mice versus worm-free controls. Similar results were observed even after two additional boosts, while clearance of worms with anthelmintic treatment 3 weeks prior to primary immunization significantly reversed the inhibitory effect of helminth infection. In contrast, helminth infection had no inhibitory effect on immunization with irradiated sporozoites. Mean anti-CSP antibody responses were similar between H. polygyrus-infected and worm-free control mice following immunization with a single dose (65,000 sporozoites) of live radiation attenuated (irradiated) Plasmodium yoelii sporozoites (17X, non-lethal strain), and protection upon sporozoite challenge was equivalent between groups. These results indicate that helminth infection may adversely affect certain anti-malarial vaccine strategies, and highlight the importance of these interactions for malaria vaccine development.
Collapse
Affiliation(s)
- Gregory S Noland
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
145
|
Rausch S, Huehn J, Loddenkemper C, Hepworth MR, Klotz C, Sparwasser T, Hamann A, Lucius R, Hartmann S. Establishment of nematode infection despite increased Th2 responses and immunopathology after selective depletion of Foxp3+ cells. Eur J Immunol 2010; 39:3066-77. [PMID: 19750483 DOI: 10.1002/eji.200939644] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Here, we show that Treg limit intestinal pathology during nematode infection and that they control the onset and magnitude of the anti-parasitic Th Th2 response. Using mice expressing the diphtheria toxin receptor under the control of the foxp3 locus, we removed Foxp3(+) Treg during the early phase of infection with Heligmosomoides polygyrus bakeri. Depletion of Treg in infected animals did not affect adult worm burden, but led to increased pathology at the site of infection. Infected, depleted mice displayed higher frequencies of activated CD4(+) T cells and increased levels of the Th2 cytokines IL-4 and IL-13. The stronger parasite-specific Th2 response was accompanied by higher levels of IL-10. Only a moderate change in Th1 (IFN-gamma) reactivity was detected in worm-infected, Treg-depleted mice. Furthermore, we detected an accelerated onset of parasite-specific Th2 and IL-10 responses in the transient absence of Foxp3(+) Treg. However, adult worm burdens were not affected by the increased Th2-reactivity in Treg-depleted mice. Hence, our data show that Treg restrict the onset and strength of Th2 responses during intestinal worm infection, while increasing primary Th2 responses does not necessarily lead to killing of larvae or accelerated expulsion of adult worms.
Collapse
Affiliation(s)
- Sebastian Rausch
- Department of Molecular Parasitology, Humboldt-University of Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Erdman SE, Rao VP, Olipitz W, Taylor CL, Jackson EA, Levkovich T, Lee CW, Horwitz BH, Fox JG, Ge Z, Poutahidis T. Unifying roles for regulatory T cells and inflammation in cancer. Int J Cancer 2010; 126:1651-65. [PMID: 19795459 DOI: 10.1002/ijc.24923] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Activities of CD4(+) regulatory (T(REG)) cells restore immune homeostasis during chronic inflammatory disorders. Roles for T(REG) cells in inflammation-associated cancers, however, are paradoxical. It is widely believed that T(REG) function in cancer mainly to suppress protective anticancer responses. However, we demonstrate here that T(REG) cells also function to reduce cancer risk throughout the body by efficiently downregulating inflammation arising from the gastrointestinal (GI) tract. Building on a "hygiene hypothesis" model in which GI infections lead to changes in T(REG) that reduce immune-mediated diseases, here we show that gut bacteria-triggered T(REG) may function to inhibit cancer even in extraintestinal sites. Ability of bacteria-stimulated T(REG) to suppress cancer depends on interleukin (IL)-10, which serves to maintain immune homeostasis within bowel and support a protective antiinflammatory T(REG) phenotype. However, under proinflammatory conditions, T(REG) may fail to provide antiinflammatory protection and instead contribute to a T helper (Th)-17-driven procarcinogenic process; a cancer state that is reversible by downregulation of inflammation. Consequently, hygienic individuals with a weakened IL-10 and T(REG)-mediated inhibitory loop are highly susceptible to the carcinogenic consequences of elevated IL-6 and IL-17 and show more frequent inflammation-associated cancers. Taken together, these data unify seemingly divergent disease processes such as autoimmunity and cancer and help explain the paradox of T(REG) and inflammation in cancer. Enhancing protective T(REG) functions may promote healthful longevity and significantly reduce risk of cancer.
Collapse
MESH Headings
- Adenomatous Polyposis Coli Protein/physiology
- Animals
- Blotting, Western
- Cytokines/genetics
- Cytokines/metabolism
- DNA-Binding Proteins/physiology
- Disease Models, Animal
- Female
- Flow Cytometry
- Helicobacter Infections/immunology
- Helicobacter Infections/microbiology
- Helicobacter Infections/prevention & control
- Helicobacter hepaticus/pathogenicity
- Immunoenzyme Techniques
- Inflammation/immunology
- Inflammation/microbiology
- Inflammation/prevention & control
- Interleukin-10/physiology
- Mammary Neoplasms, Animal/immunology
- Mammary Neoplasms, Animal/microbiology
- Mammary Neoplasms, Animal/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, ErbB-2/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Regulatory/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Abstract
Gastric cancer is the second most common cause of cancer death worldwide. A large body of evidence supports a causal role of Helicobacter pylori in the majority of gastric malignancies. Great strides have been made in understanding the pathogenesis of this relationship, but much remains to be learned. Moreover, because of the high prevalence of infection, the lack of definitive trials, and the challenges of H. pylori treatment, there remains no consensus on the role of routine screening and treatment of this infection to prevent cancer. This article reviews the current knowledge on H. pylori and gastric cancer and presents some of the clinical and public health challenges associated with this pathogen.
Collapse
Affiliation(s)
- V Herrera
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | |
Collapse
|
148
|
El-Masry S, El-Shahat M, Badra G, Aboel-Nour MF, Lotfy M. Helicobacter pylori and Hepatitis C Virus Coinfection in Egyptian Patients. J Glob Infect Dis 2010; 2:4-9. [PMID: 20300411 PMCID: PMC2840963 DOI: 10.4103/0974-777x.59244] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
INTRODUCTION Chronic hepatitis C virus (HCV) infection is a leading cause of end-stage liver disease worldwide. It has been shown that Helicobacter pylori (H. pylori) plays an important role in chronic gastritis, peptic ulcer disease and gastric malignancies, and its eradication has been advocated. The association between H. pylori infection and liver cirrhosis in patients with hepatitis C virus has been documented in different parts of the world; nevertheless, no conclusive data is available in Egypt. MATERIALS AND METHODS In the present study, the status of H. pylori infection was sought in 90 patients with chronic HCV infection and in 66 HCV-free healthy controls. RESULTS The study showed that the H. pylori positivity was increased significantly (P = 0.03) in the HCV-infected patients when compared to that in healthy controls, where H. pylori infection was found in 50 (55.6%) out of 90 of the HCV-infected patients versus 26 (39.4%) out of 66 of the healthy controls. In HCV-infected patients, the prevalence of H. pylori infection was increased significantly (P = 0.04) from chronic active hepatitis to cirrhosis. H. pylori infection was present in 6/18 (33.3%), 10/21 (47.6%), 16/27 (59.3%), 18/24 (75.0%) patients with chronic active hepatitis, Child-Pugh score A, Child-Pugh score B and Child-Pugh score C, respectively. More importantly, the prevalence of H. pylori infection in HCV-infected patients was increased very significantly (P = 0.003) with increasing Meld (model for end-stage liver disease) score. The prevalence of H. pylori was documented in 9/28 (32.1%) patients with Meld score >10 and in 41/62 (66.1%) patients with Meld score >10. CONCLUSION It may be stated that our results collectively reflect a remarkable increase in H. pylori prevalence with advancing hepatic lesions, and the eradication treatment may prove beneficial in those patients with chronic hepatitis C.
Collapse
Affiliation(s)
- Samir El-Masry
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, Minufiya University, Sadat City, Minufiya, Egypt, Saudi Arabia
| | - Mohamed El-Shahat
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, Minufiya University, Sadat City, Minufiya, Egypt, Saudi Arabia
| | - Gamal Badra
- Department of Hepatology, National Liver Institute, Minufiya University, Minufiya, Egypt, Saudi Arabia
| | - Mohamed F Aboel-Nour
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura, Egypt, Saudi Arabia
- Department of Applied Medical Sciences, Jouf University, Qurayat, Saudi Arabia
| | - Mahmoud Lotfy
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, Minufiya University, Sadat City, Minufiya, Egypt, Saudi Arabia
- Department of Applied Medical Sciences, Jouf University, Qurayat, Saudi Arabia
| |
Collapse
|
149
|
Erdman SE, Poutahidis T. Roles for inflammation and regulatory T cells in colon cancer. Toxicol Pathol 2009; 38:76-87. [PMID: 20019355 DOI: 10.1177/0192623309354110] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Risk for developing cancer rises substantially as a result of poorly regulated inflammatory responses to pathogenic bacterial infections. Anti-inflammatory CD4(+) regulatory cells (T(REG)) function to restore immune homeostasis during chronic inflammatory disorders. It seems logical that T(REG) cells would function to reduce risk of inflammation-associated cancer in the bowel by down-regulating inflammation. It is widely believed, however, that T(REG) function in cancer mainly to suppress protective anticancer inflammatory responses. Thus roles for inflammation, T(REG) cells, and gut bacteria in cancer are paradoxical and are the subject of controversy. Our accumulated data build upon the "hygiene hypothesis" model in which gastrointestinal (GI) infections lead to changes in T(REG) that reduce inflammation-associated diseases. Ability of T(REG) to inhibit or suppress cancer depends upon gut bacteria and IL-10, which serve to maintain immune balance and a protective anti-inflammatory T(REG) phenotype. However, under poorly regulated pro-inflammatory conditions, T(REG) fail to inhibit and may instead contribute to a T helper (Th)-17-driven procarcinogenic process, a cancer state that is reversible by down-regulation of inflammation and interleukin (IL)-6. Consequently, hygienic individuals with a weakened IL-10- and T(REG)-mediated inhibitory loop are highly susceptible to the carcinogenic consequences of elevated inflammation and show more frequent inflammation-associated cancers. Taken together, these data help explain the paradox of inflammation and T(REG) in cancer and indicate that targeted stimulation of T(REG) may promote health and significantly reduce risk of cancer.
Collapse
Affiliation(s)
- Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | | |
Collapse
|
150
|
Wang SSW, Asfaha S, Okumura T, Betz KS, Muthupalani S, Rogers AB, Tu S, Takaishi S, Jin G, Yang X, Wu DC, Fox JG, Wang TC. Fibroblastic colony-forming unit bone marrow cells delay progression to gastric dysplasia in a helicobacter model of gastric tumorigenesis. Stem Cells 2009; 27:2301-11. [PMID: 19591219 DOI: 10.1002/stem.165] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bone marrow mesenchymal stem cells (MSCs) have been shown to have immune modulatory effects. Despite efforts to identify these cells in vivo, to date, MSCs have been defined mainly by their in vitro cell characteristics. Here, we show that Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells make up approximately 0.5%-1% of murine whole bone marrow cells and yield nearly an equal amount of fibroblastic colony-forming units (CFU-F) as whole bone marrow. After transplantation into lethally irradiated recipients, Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells engrafted in the bone marrow long-term and demonstrated characteristics of MSCs, including capacity to differentiate into osteoblasts and adipocytes. To examine whether Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells have immune modulatory effects, in vitro coculture with activated CD4+ T-cells resulted in decreased Th17 cell differentiation by Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells. Furthermore, serial infusions with Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells reduced the progression to low-grade gastric dysplasia in mice infected with chronic Helicobacter felis (p = .038). This correlated with reduced gastric interleukin (IL)-17F, IL-22, and ROR-gammat gene expression in responding mice (p < .05). These data suggest that bone marrow derived Lin(-)CD44(hi)Sca1(-)cKit+CD34(-) cells have characteristics of MSCs and reduce progression of early gastric tumorigenesis induced by chronic H. felis infection. The prevention of dysplastic changes may occur through inhibition of Th17-dependent pathways.
Collapse
Affiliation(s)
- Sophie S W Wang
- Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|