Predictive computational modeling of the mucosal immune responses during Helicobacter pylori infection

Adria Carbo, Josep Bassaganya-Riera, Mireia Pedragosa, Monica Viladomiu, Madhav Marathe, Stephen Eubank, Katherine Wendelsdorf, Keith Bisset, Stefan Hoops, Xinwei Deng, Maksudul Alam, Barbara Kronsteiner, Yongguo Mei, Raquel Hontecillas, Adria Carbo, Josep Bassaganya-Riera, Mireia Pedragosa, Monica Viladomiu, Madhav Marathe, Stephen Eubank, Katherine Wendelsdorf, Keith Bisset, Stefan Hoops, Xinwei Deng, Maksudul Alam, Barbara Kronsteiner, Yongguo Mei, Raquel Hontecillas

Abstract

T helper (Th) cells play a major role in the immune response and pathology at the gastric mucosa during Helicobacter pylori infection. There is a limited mechanistic understanding regarding the contributions of CD4+ T cell subsets to gastritis development during H. pylori colonization. We used two computational approaches: ordinary differential equation (ODE)-based and agent-based modeling (ABM) to study the mechanisms underlying cellular immune responses to H. pylori and how CD4+ T cell subsets influenced initiation, progression and outcome of disease. To calibrate the model, in vivo experimentation was performed by infecting C57BL/6 mice intragastrically with H. pylori and assaying immune cell subsets in the stomach and gastric lymph nodes (GLN) on days 0, 7, 14, 30 and 60 post-infection. Our computational model reproduced the dynamics of effector and regulatory pathways in the gastric lamina propria (LP) in silico. Simulation results show the induction of a Th17 response and a dominant Th1 response, together with a regulatory response characterized by high levels of mucosal Treg) cells. We also investigated the potential role of peroxisome proliferator-activated receptor γ (PPARγ) activation on the modulation of host responses to H. pylori by using loss-of-function approaches. Specifically, in silico results showed a predominance of Th1 and Th17 cells in the stomach of the cell-specific PPARγ knockout system when compared to the wild-type simulation. Spatio-temporal, object-oriented ABM approaches suggested similar dynamics in induction of host responses showing analogous T cell distributions to ODE modeling and facilitated tracking lesion formation. In addition, sensitivity analysis predicted a crucial contribution of Th1 and Th17 effector responses as mediators of histopathological changes in the gastric mucosa during chronic stages of infection, which were experimentally validated in mice. These integrated immunoinformatics approaches characterized the induction of mucosal effector and regulatory pathways controlled by PPARγ during H. pylori infection affecting disease outcomes.

Conflict of interest statement

Competing Interests: The authors state that Josep Bassaganya-Riera is an Academic Editor for PLOS ONE. This does not alter the authors′ adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Network model of the mucosal…
Figure 1. Network model of the mucosal immune responses during Helicobacter pylori infection.
Systems Biology Markup Language (SBML)-compliant network of the interactions between H. pylori and cells participating in the innate and adaptive immune response such as macrophages (M1 and M2), dendritic cells (tDC and eDC), epithelial cells (E) and CD4+ T cell subsets (Th1, Th17, iTreg) in the gastric lumen, the epithelium, lamina propria (LP) and the gastric lymph nodes (GLN).
Figure 2. Effector and regulatory CD4+ T…
Figure 2. Effector and regulatory CD4+ T cell subsets modulate the immune responses during Helicobacter pylori infection.
(A) In silico time-course experiment performed with a challenge of 5×107 colony forming units of initial H. pylori injected in the mathematical model, showing differences in numbers of gastric lamina propria (LP) CD4+ T cell subsets over time. (B) Equilibrium constant regulating CD4+ T cell gastric lymph nodes (GLN) differentiation in our computational model. (C, D) Flow cytometry analysis results showing differences in the percentages of regulatory T (Treg) cells in spleen and GLN. (E) Flow cytometric analysis showing differences in the expression of IFNγ+ Tbet+ CD4+ T (Th1) cells in the spleen at day 30 post-infection. (F, G) Histopathological analysis on the gastric mucosa showed lesions consistent with H. pylori infection. Mouse stomachs had increased leukocyte infiltration in the LP and gastric mucosal thickening due to epithelial cell proliferation.
Figure 3. In silico dynamics of gastric…
Figure 3. In silico dynamics of gastric mucosal T cell subset in wild-type and T cell-specific peroxisome proliferator-activated receptor γ (PPARγ) knockout mice following infection with Helicobacter pylori.
Time-course experiments following infection with 5×107 colony-forming units (CFU) of H. pylori to determine dynamics on CD4+ T cell phenotypes. Blue lines represent wild type mice and violet lines represent T cell-specific PPARγ knockout mice. T helper (Th) 1 (A), Th17 (B), induced regulatory T cell (iTreg) (C), effector dendritic cells (D), tolerogenic dendritic cells (E), M1 macrophages (F) and M2 macrophages (G) are illustrated.
Figure 4. Enteric Immunity Simulator (ENISI) output…
Figure 4. Enteric Immunity Simulator (ENISI) output results and assessment of the role of the Peroxisome Proliferator Activated Receptor γ (PPARγ) in both the myeloid and T cell subset modulated T cell responses after Helicobacter pylori infection in silico in the gastric lamina propria (LP) and gastric lymph nodes (GLN).
The H. pylori ABM was run as a time-course for 60 days. Model parameters were changed to simulate myeloid or T cell-specific PPARγ knockout systems as described in Table S2. Dynamical variation of Th1 (Figure 6A, 6G) as well as Th17 (Figure 6B, 6H) and regulatory T cells (Figure 6C, 6I) changing over time were plotted. A functional T-test was used with 95% confidence interval to create statistics assessing differences in the myeloid and T cell specific PPARγ knock-out for Th1 (Figure 6D, 6J), Th17 (Figure 6E, 6K) and Treg (Figure 6F, 6L). A threshold value representing the critical value of significance vertically divides the plot into two parts, showing significant differences above the threshold. Data were obtained in 15 runs of the simulation for each different genotype.
Figure 5. Sensitivity analysis of factors involved…
Figure 5. Sensitivity analysis of factors involved in gastric inflammatory lesion formation following Helicobacter pylori infection.
Healthy epithelial cells changing state into pro-inflammatory epithelial cells, thereby contributing to the formation of gastric lesions. (A) Differential time-dependent patterns of lesion formation in the early, meridian and chronic-late stage of infection. (B) ODE-based deterministic sensitivity analysis on pro-inflammatory epithelial cells, as variables, and its formation at day 60 post-infection using a delta factor of 0.001 with a delta minimum of 1×10−12. (C) Flow cytometric analysis showing differences in the expression of CD4+ IL-17A+ cells in the gastric lamina propria after H. pylori infection. (D) Flow cytometric analysis showing differences in the expression of CD4+ IFNγ+ cells in the gastric lamina propria after H. pylori infection. (E) Cartoon model representation of the effect of DC activation, T cell expansion and macrophage differentiation on the formation of histopathological lesions in the gastric lamina propria (LP) during H. pylori infection.
Figure 6. Histopathological assessment of the gastric…
Figure 6. Histopathological assessment of the gastric mucosa of mice after Helicobacter pylori infection.
Representative photomicrographs of stomachs from either non infected or H. pylori-infected mice following administration of PBS or metronidazole treatment. Original magnification 40×.

References

    1. Atherton JC, Blaser MJ (2009) Coadaptation of Helicobacter pylori and humans: ancient history, modern implications. J Clin Invest 119: 2475–2487.
    1. Hitzler I, Kohler E, Engler DB, Yazgan AS, Muller A (2012) The role of Th cell subsets in the control of Helicobacter infections and in T cell-driven gastric immunopathology. Front Immunol 3: 142.
    1. Karttunen R, Karttunen T, Ekre HP, MacDonald TT (1995) Interferon gamma and interleukin 4 secreting cells in the gastric antrum in Helicobacter pylori positive and negative gastritis. Gut 36: 341–345.
    1. Bamford KB, Fan X, Crowe SE, Leary JF, Gourley WK, et al. (1998) Lymphocytes in the human gastric mucosa during Helicobacter pylori have a T helper cell 1 phenotype. Gastroenterology 114: 482–492.
    1. Smythies LE, Waites KB, Lindsey JR, Harris PR, Ghiara P, et al. (2000) Helicobacter pylori-induced mucosal inflammation is Th1 mediated and exacerbated in IL-4, but not IFN-gamma, gene-deficient mice. J Immunol 165: 1022–1029.
    1. Afkarian M, Sedy JR, Yang J, Jacobson NG, Cereb N, et al. (2002) T-bet is a STAT1-induced regulator of IL-12R expression in naive CD4+ T cells. Nat Immunol 3: 549–557.
    1. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, et al. (2006) Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441: 235–238.
    1. Philipson CW, Bassaganya-Riera J, Viladomiu M, Pedragosa M, Guerrant RL, et al. (2013) The role of peroxisome proliferator-activated receptor gamma in immune responses to enteroaggregative Escherichia coli infection. PLoS One 8: e57812.
    1. Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joannopoulos K, et al. (2006) Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med 203: 2271–2279.
    1. Marson A, Kretschmer K, Frampton GM, Jacobsen ES, Polansky JK, et al. (2007) Foxp3 occupancy and regulation of key target genes during T-cell stimulation. Nature 445: 931–935.
    1. Ando T, Goto Y, Ishiguro K, Maeda O, Watanabe O, et al. (2007) The interaction of host genetic factors and Helicobacter pylori infection. Inflammopharmacology 15: 10–14.
    1. Hontecillas R, Bassaganya-Riera J (2007) Peroxisome proliferator-activated receptor gamma is required for regulatory CD4+ T cell-mediated protection against colitis. J Immunol 178: 2940–2949.
    1. Bassaganya-Riera J, Dominguez-Bello MG, Kronsteiner B, Carbo A, Lu P, et al... (2012) Helicobacter pylori colonization ameliorates glucose homeostasis in mice through a PPAR gamma-dependent mechanism. PLoS One In Press.
    1. Bazargani A, Khoramrooz SS, Kamali-Sarvestani E, Taghavi SA, Saberifiroozi M (2010) Association between peroxisome proliferator-activated receptor-gamma gene polymorphism (Pro12Ala) and Helicobacter pylori infection in gastric carcinogenesis. Scand J Gastroenterol 45: 1162–1167.
    1. Yao L, Liu F, Sun L, Wu H, Guo C, et al. (2010) Upregulation of PPARgamma in tissue with gastric carcinoma. Hybridoma (Larchmt) 29: 341–343.
    1. Konturek PC, Kania J, Kukharsky V, Raithel M, Ocker M, et al. (2004) Implication of peroxisome proliferator-activated receptor gamma and proinflammatory cytokines in gastric carcinogenesis: link to Helicobacter pylori-infection. J Pharmacol Sci 96: 134–143.
    1. Son SH, Kim HK, Ji JS, Cho YS, Kim SS, et al. (2007) [Expression of peroxisome proliferator-activated receptor (PPAR) gamma in Helicobacter pylori-infected gastric epithelium]. Korean J Gastroenterol 49: 72–78.
    1. Viladomiu M, Hontecillas R, Pedragosa M, Carbo A, Hoops S, et al. (2012) Modeling the role of peroxisome proliferator-activated receptor gamma and microRNA-146 in mucosal immune responses to Clostridium difficile. PLoS One 7: e47525.
    1. Carbo A, Hontecillas R, Kronsteiner B, Viladomiu M, Pedragosa M, et al. (2013) Systems modeling of molecular mechanisms controlling cytokine-driven CD4+ T cell differentiation and phenotype plasticity. PLoS Comput Biol 9: e1003027.
    1. Lundgren A, Trollmo C, Edebo A, Svennerholm AM, Lundin BS (2005) Helicobacter pylori-specific CD4+ T cells home to and accumulate in the human Helicobacter pylori-infected gastric mucosa. Infect Immun 73: 5612–5619.
    1. Tan S, Tompkins LS, Amieva MR (2009) Helicobacter pylori usurps cell polarity to turn the cell surface into a replicative niche. PLoS Pathog 5: e1000407.
    1. Naumann M, Crabtree JE (2004) Helicobacter pylori-induced epithelial cell signalling in gastric carcinogenesis. Trends Microbiol 12: 29–36.
    1. Andres S, Schmidt HM, Mitchell H, Rhen M, Maeurer M, et al. (2011) Helicobacter pylori defines local immune response through interaction with dendritic cells. FEMS Immunol Med Microbiol 61: 168–178.
    1. Zhang M, Liu M, Luther J, Kao JY (2010) Helicobacter pylori directs tolerogenic programming of dendritic cells. Gut Microbes 1: 325–329.
    1. Quiding-Jarbrink M, Raghavan S, Sundquist M (2010) Enhanced M1 macrophage polarization in human helicobacter pylori-associated atrophic gastritis and in vaccinated mice. PLoS One 5: e15018.
    1. Bimczok D, Grams JM, Stahl RD, Waites KB, Smythies LE, et al. (2011) Stromal regulation of human gastric dendritic cells restricts the Th1 response to Helicobacter pylori. Gastroenterology 141: 929–938.
    1. Wong BL, Zhu SL, Huang XR, Ma J, Xia HH, et al. (2009) Essential role for macrophage migration inhibitory factor in gastritis induced by Helicobacter pylori. Am J Pathol 174: 1319–1328.
    1. Zhuang Y, Shi Y, Liu XF, Zhang JY, Liu T, et al. (2011) Helicobacter pylori-infected macrophages induce Th17 cell differentiation. Immunobiology 216: 200–207.
    1. Hoops S, Sahle S, Gauges R, Lee C, Pahle J, et al. (2006) COPASI–a COmplex PAthway SImulator. Bioinformatics 22: 3067–3074.
    1. Wendelsdorf K, Alam M, Bassaganya-Riera J, Bisset K, Eubank S, et al. (2012) ENteric Immunity SImulator: A tool for in silico study of gastroenteric infections. IEEE Transactions on NanoBioScience 11: 273–288.
    1. Funahashi A, Tanimura N, Morohashi M, Kitano H (2003) CellDesigner: a process diagram editor for gene-regulatory and biochemical networks. BIOSILICO 1: 159–162.
    1. Iwasaki A (2007) Mucosal dendritic cells. Annu Rev Immunol 25: 381–418.
    1. Ng SC, Kamm MA, Stagg AJ, Knight SC (2010) Intestinal dendritic cells: their role in bacterial recognition, lymphocyte homing, and intestinal inflammation. Inflamm Bowel Dis 16: 1787–1807.
    1. Littman DR, Rudensky AY (2010) Th17 and regulatory T cells in mediating and restraining inflammation. Cell 140: 845–858.
    1. Kennedy J (1997) The particle swarm: social adaptation of knowledge. IEEE Congress on Evolutionary Computation: 303–308.
    1. Hoops S, Sahle S, Gauges R, Lee C, Pahle J, et al. (2006) COPASI–a complex pathway simulator. Bioinformatics 22: 3067–3074.
    1. Spiegelman BM (1998) PPARgamma in monocytes: less pain, any gain? Cell 93: 153–155.
    1. Klotz L, Burgdorf S, Dani I, Saijo K, Flossdorf J, et al. (2009) The nuclear receptor PPAR gamma selectively inhibits Th17 differentiation in a T cell-intrinsic fashion and suppresses CNS autoimmunity. J Exp Med 206: 2079–2089.
    1. Mansen A, Guardiola-Diaz H, Rafter J, Branting C, Gustafsson JA (1996) Expression of the peroxisome proliferator-activated receptor (PPAR) in the mouse colonic mucosa. Biochem Biophys Res Commun 222: 844–851.
    1. Wohlfert EA, Nichols FC, Nevius E, Clark RB (2007) Peroxisome proliferator-activated receptor gamma (PPARgamma) and immunoregulation: enhancement of regulatory T cells through PPARgamma-dependent and -independent mechanisms. J Immunol 178: 4129–4135.
    1. Adria Carbo RH, Kronsteiner B, Viladomiu M, Pedragosa M, Lu P, et al... (2013) Systems modeling of molecular mechanisms controlling cytokine-driven CD4+ T cell differentiation and phenotype plasticity. PLoS Comput Biol In Press.
    1. Bisset KR, Alam MM, Bassaganya-Riera J, Carbo A, Eubank S, et al. High-Performance Interaction-Based Simulation of Gut Immunopathologies with ENteric Immunity Simulator (ENISI); 2012. IEEE Computer Society. 48–59.
    1. Olokoba AB, Obateru OA, Bojuwoye MO (2013) Helicobacter pylori eradication therapy: A review of current trends. Niger Med J 54: 1–4.
    1. Parsonnet J, Hansen S, Rodriguez L, Gelb AB, Warnke RA, et al. (1994) Helicobacter pylori infection and gastric lymphoma. N Engl J Med 330: 1267–1271.
    1. Parsonnet J, Isaacson PG (2004) Bacterial infection and MALT lymphoma. N Engl J Med 350: 213–215.
    1. Correa P, Houghton J (2007) Carcinogenesis of Helicobacter pylori. Gastroenterology 133: 659–672.
    1. Blaser MJ (2008) Disappearing microbiota: Helicobacter pylori protection against esophageal adenocarcinoma. Cancer Prev Res (Phila Pa) 1: 308–311.
    1. Vieth M, Masoud B, Meining A, Stolte M (2000) Helicobacter pylori infection: protection against Barrett’s mucosa and neoplasia? Digestion 62: 225–231.
    1. Vaezi MF, Falk GW, Peek RM, Vicari JJ, Goldblum JR, et al. (2000) CagA-positive strains of Helicobacter pylori may protect against Barrett’s esophagus. Am J Gastroenterol 95: 2206–2211.
    1. Chow WH, Blaser MJ, Blot WJ, Gammon MD, Vaughan TL, et al. (1998) An inverse relation between cagA+ strains of Helicobacter pylori infection and risk of esophageal and gastric cardia adenocarcinoma. Cancer Res 58: 588–590.
    1. Blaser MJ, Chen Y, Reibman J (2008) Does Helicobacter pylori protect against asthma and allergy? Gut.
    1. Chen Y, Blaser MJ (2007) Inverse associations of Helicobacter pylori with asthma and allergy. Arch Intern Med 167: 821–827.
    1. Lang L (2007) Childhood acquisition of Helicobacter pylori linked to reduced asthma and allergy risk. Gastroenterology 133: 6.
    1. McCune A, Lane A, Murray L, Harvey I, Nair P, et al. (2003) Reduced risk of atopic disorders in adults with Helicobacter pylori infection. Eur J Gastroenterol Hepatol 15: 637–640.
    1. Harris PR, Wright SW, Serrano C, Riera F, Duarte I, et al. (2008) Helicobacter pylori gastritis in children is associated with a regulatory T-cell response. Gastroenterology 134: 491–499.
    1. Jang TJ (2010) The number of Foxp3-positive regulatory T cells is increased in Helicobacter pylori gastritis and gastric cancer. Pathol Res Pract 206: 34–38.
    1. Esplugues E, Huber S, Gagliani N, Hauser AE, Town T, et al. (2011) Control of TH17 cells occurs in the small intestine. Nature 475: 514–518.
    1. Sayi A, Kohler E, Hitzler I, Arnold I, Schwendener R, et al. (2009) The CD4+ T cell-mediated IFN-gamma response to Helicobacter infection is essential for clearance and determines gastric cancer risk. J Immunol 182: 7085–7101.
    1. Shi Y, Liu XF, Zhuang Y, Zhang JY, Liu T, et al. (2010) Helicobacter pylori-induced Th17 responses modulate Th1 cell responses, benefit bacterial growth, and contribute to pathology in mice. J Immunol 184: 5121–5129.
    1. Algood HM, Allen SS, Washington MK, Peek RM Jr, Miller GG, et al. (2009) Regulation of gastric B cell recruitment is dependent on IL-17 receptor A signaling in a model of chronic bacterial infection. J Immunol 183: 5837–5846.
    1. O’Connor W Jr, Zenewicz LA, Flavell RA (2010) The dual nature of T(H)17 cells: shifting the focus to function. Nat Immunol 11: 471–476.
    1. Quiding-Jarbrink M, Lundin BS, Lonroth H, Svennerholm AM (2001) CD4+ and CD8+ T cell responses in Helicobacter pylori-infected individuals. Clin Exp Immunol 123: 81–87.
    1. Krakowka S, Ringler SS, Eaton KA, Green WB, Leunk R (1996) Manifestations of the local gastric immune response in gnotobiotic piglets infected with Helicobacter pylori. Vet Immunol Immunopathol 52: 159–173.
    1. Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK (1998) The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature 391: 79–82.
    1. Wu L, Yan C, Czader M, Foreman O, Blum JS, et al. (2012) Inhibition of PPARgamma in myeloid-lineage cells induces systemic inflammation, immunosuppression, and tumorigenesis. Blood 119: 115–126.
    1. Abdullah Z, Geiger S, Nino-Castro A, Bottcher JP, Muraliv E, et al. (2012) Lack of PPARgamma in myeloid cells confers resistance to Listeria monocytogenes infection. PLoS One 7: e37349.
    1. Zhang MA, Rego D, Moshkova M, Kebir H, Chruscinski A, et al. (2012) Peroxisome proliferator-activated receptor (PPAR)alpha and -gamma regulate IFNgamma and IL-17A production by human T cells in a sex-specific way. Proc Natl Acad Sci U S A 109: 9505–9510.
    1. Bassaganya-Riera J, Viladomiu M, Pedragosa M, De Simone C, Hontecillas R (2012) Immunoregulatory mechanisms underlying prevention of colitis-associated colorectal cancer by probiotic bacteria. PLoS One 7: e34676.
    1. Klotz L, Knolle P (2011) Nuclear receptors: TH17 cell control from within. FEBS Lett 585: 3764–3769.
    1. Lei J, Hasegawa H, Matsumoto T, Yasukawa M (2010) Peroxisome proliferator-activated receptor alpha and gamma agonists together with TGF-beta convert human CD4+CD25− T cells into functional Foxp3+ regulatory T cells. J Immunol 185: 7186–7198.
    1. Hoechst B, Gamrekelashvili J, Manns MP, Greten TF, Korangy F (2011) Plasticity of human Th17 cells and iTregs is orchestrated by different subsets of myeloid cells. Blood 117: 6532–6541.
    1. Kryczek I, Wu K, Zhao E, Wei S, Vatan L, et al. (2011) IL-17+ regulatory T cells in the microenvironments of chronic inflammation and cancer. J Immunol 186: 4388–4395.
    1. Nagai S, Mimuro H, Yamada T, Baba Y, Moro K, et al. (2007) Role of Peyer’s patches in the induction of Helicobacter pylori-induced gastritis. Proc Natl Acad Sci U S A 104: 8971–8976.

Source: PubMed

3
Prenumerera