Gut microbiota is a key modulator of insulin resistance in TLR 2 knockout mice

Andréa M Caricilli, Paty K Picardi, Lélia L de Abreu, Mirian Ueno, Patrícia O Prada, Eduardo R Ropelle, Sandro Massao Hirabara, Ângela Castoldi, Pedro Vieira, Niels O S Camara, Rui Curi, José B Carvalheira, Mário J A Saad, Andréa M Caricilli, Paty K Picardi, Lélia L de Abreu, Mirian Ueno, Patrícia O Prada, Eduardo R Ropelle, Sandro Massao Hirabara, Ângela Castoldi, Pedro Vieira, Niels O S Camara, Rui Curi, José B Carvalheira, Mário J A Saad

Abstract

Environmental factors and host genetics interact to control the gut microbiota, which may have a role in the development of obesity and insulin resistance. TLR2-deficient mice, under germ-free conditions, are protected from diet-induced insulin resistance. It is possible that the presence of gut microbiota could reverse the phenotype of an animal, inducing insulin resistance in an animal genetically determined to have increased insulin sensitivity, such as the TLR2 KO mice. In the present study, we investigated the influence of gut microbiota on metabolic parameters, glucose tolerance, insulin sensitivity, and signaling of TLR2-deficient mice. We investigated the gut microbiota (by metagenomics), the metabolic characteristics, and insulin signaling in TLR2 knockout (KO) mice in a non-germ free facility. Results showed that the loss of TLR2 in conventionalized mice results in a phenotype reminiscent of metabolic syndrome, characterized by differences in the gut microbiota, with a 3-fold increase in Firmicutes and a slight increase in Bacteroidetes compared with controls. These changes in gut microbiota were accompanied by an increase in LPS absorption, subclinical inflammation, insulin resistance, glucose intolerance, and later, obesity. In addition, this sequence of events was reproduced in WT mice by microbiota transplantation and was also reversed by antibiotics. At the molecular level the mechanism was unique, with activation of TLR4 associated with ER stress and JNK activation, but no activation of the IKKβ-IκB-NFκB pathway. Our data also showed that in TLR2 KO mice there was a reduction in regulatory T cell in visceral fat, suggesting that this modulation may also contribute to the insulin resistance of these animals. Our results emphasize the role of microbiota in the complex network of molecular and cellular interactions that link genotype to phenotype and have potential implications for common human disorders involving obesity, diabetes, and even other immunological disorders.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. Metabolic parameters of TLR2 knockout…
Figure 1. Metabolic parameters of TLR2 knockout (TLR2−/−) and WT mice during 16 wk.
(A) Weight gain after 16 wk. (B) Food intake after 8 and 16 wk. (C) Epididymal fat pad weight after 8 and 16 wk. (D) WT and TLR2−/− mice after 20 wk. (E) Glucose tolerance test. (F) Serum insulin concentration. (G) Glucose uptake obtained from euglycaemic hyperinsunaemic clamp. (H) Oxygen consumption and (I) respiratory exchange rate. (J) UCP-1 expression in the brown adipose tissue. Equal protein loading in the gel was confirmed by reblotting the membrane with an anti-β-actin antibody (J, lower panel). All evaluations were made with mice on standard chow. Data are presented as means ± S.E.M from six to eight mice per group from experiments that were repeated at least three times. *p<0.05 between TLR2−/− mice and their controls.
Figure 2. TLR2 KO mice exhibit taxonomical…
Figure 2. TLR2 KO mice exhibit taxonomical alterations in gut microbiota.
Untreated WT (A) and TLR2 knockout (TLR2−/−) mice (B) stools were analyzed via 16S rRNA analysis. Data are presented from six to eight mice per group from experiments that were repeated at least three times. All evaluations were made with mice on standard chow.
Figure 3. Measurements of cytokines, adipokines, and…
Figure 3. Measurements of cytokines, adipokines, and LPS.
Serum concentration of IL-6 (A), TNF-α (B), adiponectin (C), leptin (D), and LPS (E). Proportion of Bifidobacterium was obtained by 16S rRNA analysis of stools (F). Data are presented as means ± S.E.M from six to eight mice per group from experiments that were repeated at least three times. * p<0.05 between TLR2 KO mice and their controls; all evaluations were made with mice on standard chow.
Figure 4. Evaluation of pathways involved in…
Figure 4. Evaluation of pathways involved in the impairment of insulin signaling.
Phosphorylation of JNK in muscle (A), liver (B), and white adipose tissue (WAT) (C). Phosphorylation of PERK in muscle (D), liver (E), and WAT (F). Serine 307 phosphorylation of IRS-1 from muscle (G), liver (H), and WAT (I). Activation of TLR4 (studied by the immunoprecipitation of MyD88 and blotting with TLR4) in muscle (J), liver (K), and WAT (L). JNK, PERK, and IRS-1 protein expression in muscle, liver, and white adipose tissue (A–I, lower panels). Expression of IκB-α in muscle (M), liver (N), and WAT (O). Equal protein loading in the gel was confirmed by reblotting the membrane with an anti-β-actin antibody (M–O, lower panels). NFκB activation in muscle (P), liver (Q), and WAT (R). All evaluations were made with mice on standard chow. Data are presented as means ± S.E.M from six to eight mice per group from experiments that were repeated at least three times. * p<0.05 between TLR2−/− mice and their controls.
Figure 5. TLR2 knockout (TLR2−/−) mice present…
Figure 5. TLR2 knockout (TLR2−/−) mice present decreased insulin signaling.
Tyrosine 941 phosphorylation of the insulin receptor substrate (IRS)-1 in muscle (A), liver (B), and WAT (C). Serine phosphorylation of AKT in muscle (D), liver (E), and WAT (F). IRS-1 and AKT protein expression in muscle, liver, and white adipose tissue (A–F, lower panels). Equal protein loading in the gel was confirmed by reblotting the membrane with an anti-β-actin antibody (lower panels). All evaluations were made with mice on standard chow. Data are presented as means ± S.E.M from six to eight mice per group from experiments that were repeated at least three times. *p<0.05 between WT with and without insulin stimulus; ** p<0.05 between TLR2−/− mice and their controls with insulin stimulus.
Figure 6. Insulin sensitivity and signaling after…
Figure 6. Insulin sensitivity and signaling after treatment with selective inhibitors.
Glucose uptake obtained by the euglycaemic hyperinsulinaemic clamp from TLR2−/− mice treated or not with the drugs: SP600125 (SP), JNK inhibitor; 4-phenil butyric acid (PBA), endoplasmic reticulum stress inhibitor; TLR4 antisense oligonucleotide (ASO); (A) TAK-242, inhibitor of TLR4. (B) Serine phosphorylation of AKT after the treatment with SP600125 and PBA. (C) Serine phosphorylation of AKT after the treatment with TLR4 ASO and TAK-242. (D) Phosphorylation of JNK after the treatment with the drugs mentioned. Fasted TLR2 knockout mice and WT mice were gavaged by LPS (1.08, 10−8 g) diluted in water (100 µL) or without LPS. (E) Blood was collected from the cava vein 60 min after gavage and serum LPS was determined. (F) Zonula occludens (ZO)-1 expression in the ileum. (G) Frequency of CD4+Foxp3+ regulatory T cells in WT mice. (H) Frequency of CD4+Foxp3+ regulatory T cells in TLR2−/− mice. All evaluations were made with mice on standard chow. Data are presented as means ± S.E.M from six to eight mice per group from experiments that were repeated at least three times. * p<0.05 between WT mice with or without insulin stimulus; ** p<0.05 between WT and TLR2−/− mice with insulin stimulus ; $ p<0.05 between TLR2−/− mice with insulin stimulus, treated or not with SP; # p<0.05 between TLR2−/− mice with insulin stimulus, treated or not with PBA; § p<0.05 between TLR2−/− mice with insulin stimulus, treated or not with ASO; & p<0.05 between TLR2−/− mice with insulin stimulus, treated or not with TAK-242; ap<0.05 between WT mice with or without LPS stimulus; ° p<0.05 between WT and TLR2−/− mice with LPS stimulus.
Figure 7. Alterations in the metabolic parameters…
Figure 7. Alterations in the metabolic parameters and in insulin signaling and sensitivity after treatment with antibiotics.
(A) Epididymal fat pad weight. (B) Visceral adipose tissue weight. (C) Glucose tolerance test. (D) Oxygen consumption. (E) Glucose uptake obtained by the euglycaemic hyperinsulinaemic clamp. (F) UCP-1 expression in the brown adipose tissue. (G) Serine phosphorylation of AKT after the treatment with AB. (H) Phosphorylation of JNK after the treatment with AB. (I) Zonula occludens (ZO)-1 expression in the ileum. Equal protein loading in the gel was confirmed by reblotting the membrane with an anti-β-actin antibody (lower panels). Data are presented as means ± S.E.M from six to eight mice per group, from experiments that were repeated at least three times. All evaluations were made with mice on standard chow. # p<0.05 between WT mice with or without insulin stimulus; * p<0.05 between WT and TLR2−/− mice with insulin stimulus; ** p<0.05 between TLR2−/− and TLR2−/− treated with AB, with insulin stimulus.
Figure 8. WT mice reproduce TLR2 knockout…
Figure 8. WT mice reproduce TLR2 knockout (TLR2−/−) mice after cecal microbiota transplantation.
(A) Epididymal fat pad weight. (B) Visceral adipose tissue weight. (C) Weight gain of transplanted mice. (D) Serum glucose. (E) Glucose tolerance test. (F) Incremental area under curva (IAUC) obtained from the glucose tolerance test. (G) Oxygen consumption. (H) Glucose uptake obtained by the euglycaemic hyperinsulinaemic clamp. (I) UCP-1 expression in the brown adipose tissue. (J) Serine phosphorylation of AKT after the treatment with AB. (K) Phosphorylation of JNK after the treatment with AB. AKT and JNK protein expression in the liver of transplanted mice (J, K, lower panels). (L) Zonula occludens (ZO)-1 expression in the ileum. (M) Frequency of CD4+Foxp3+ regulatory T cells in Bacillus-associated mice. (N) Frequency of CD4+Foxp3+ regulatory T cells in Bacillus-associated mice transplanted with WT microbiota. (O) Frequency of CD4+Foxp3+ regulatory T cells in Bacillus-associated mice transplanted with TLR2−/− microbiota. Equal protein loading in the gel was confirmed by reblotting the membrane with an anti-β-actin antibody (lower panels). Data are presented as means ± S.E.M from six to eight mice per group from experiments that were repeated at least three times. All evaluations were made with mice on standard chow. * p<0.05 between Bacillus-associated mice transplanted with TLR2−/− microbiota (MA+TLR2−/−) and those transplanted with WT microbiota (MA+WT); ** p<0.05 between Bacillus-associated mice transplanted with WT microbiota (MA+WT) and Bacillus-associated mice (MA); # p<0.05 between Bacillus-associated mice with or without insulin stimulus.
Figure 9. Metabolic parameters of TLR2 KO…
Figure 9. Metabolic parameters of TLR2 KO (TLR2−/−) mice fed a high-fat diet.
(A) Weight gain after 10 wk of high-fat diet (HFD). (B) Food intake. (C) Epididymal fat pad weight. (D) Glucose tolerance test. (E) Serum insulin concentration. (F) Glucose uptake obtained from the euglycaemic hyperinsulinaemic clamp. (G) Oxygen consumption and (H) respiratory exchange rate. (I) UCP-1 expression in the brown adipose tissue. Data are presented as means ± S.E.M from six to eight mice per group from experiments that were repeated at least three times. All evaluations were made with mice on standard chow. * p<0.05 between TLR2−/− mice and their controls.
Figure 10. Insulin signaling is impaired in…
Figure 10. Insulin signaling is impaired in TLR2 knockout (TLR2−/−) mice fed on a high-fat diet.
Phosphorylation of AKT in muscle (A), liver (B), and white adipose tissue (WAT) (C). AKT protein expression in muscle, liver, and WAT (A–C, lower panels). Phosphorylation of JNK in muscle (D), liver (E), and WAT (F). JNK protein expression in muscle, liver, and WAT (D–F, lower panels). IκB-α expression in muscle (G), liver (H), and WAT (I). Equal protein loading in the gel was confirmed by reblotting the membrane with an anti-β-actin antibody (lower panels). All evaluations were made with mice on standard chow. Data are presented as means ± S.E.M from six to eight mice per group from experiments that were repeated at least three times. * p<0.05 between TLR2−/− mice and their controls, with insulin stimulus; ** p<0.05 between WT with and without insulin stimulus.

References

    1. Hossain P, Kawar B, El Nahas M. Obesity and diabetes in the developing world–a growing challenge. N Engl J Med. 2007;356:213–215.
    1. Lazar M. A. How obesity causes diabetes: not a tall tale. Science. 2005;307:373–375.
    1. Doria A, Patti M. E, Kahn C. R. The emerging genetic architecture of type 2 diabetes. Cell Metab. 2008;8:186–200.
    1. Rankinen T, Zuberi A, Chagnon Y. C, Weisnagel S. J, Argyropoulos G, et al. The human obesity gene map: the 2005 update. Obesity (Silver Spring) 2006;14:529–644.
    1. Walley A. J, Asher J. E, Froguel P. The genetic contribution to non-syndromic human obesity. Nat Rev Genet. 2009;10:431–442.
    1. Freedman A. S, Freeman G. J, Rhynhart K, Nadler L. M. Selective induction of B7/BB-1 on interferon-gamma stimulated monocytes: a potential mechanism for amplification of T cell activation through the CD28 pathway. Cell Immunol. 1991;137:429–437.
    1. Wellen K. E, Hotamisligil G. S. Inflammation, stress, and diabetes. J Clin Invest. 2005;115:1111–1119.
    1. Weisberg S. P, McCann D, Desai M, Rosenbaum M, Leibel R. L, et al. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112:1796–1808.
    1. Xu H, Barnes G. T, Yang Q, Tan G, Yang D, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112:1821–1830.
    1. Arkan M. C, Hevener A. L, Greten F. R, Maeda S, Li Z. W, et al. IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med. 2005;11:191–198.
    1. Cani P. D, Amar J, Iglesias M. A, Poggi M, Knauf C, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56:1761–1772.
    1. Cani P. D, Bibiloni R, Knauf C, Waget A, Neyrinck A. M, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57:1470–1481.
    1. Creely S. J, McTernan P. G, Kusminski C. M, Fisher M, Da Silva N. F, et al. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am J Physiol Endocrinol Metab. 2007;292:E740–E747.
    1. Chung S, Lapoint K, Martinez K, Kennedy A, Boysen Sandberg M, et al. Preadipocytes mediate lipopolysaccharide-induced inflammation and insulin resistance in primary cultures of newly differentiated human adipocytes. Endocrinology. 2006;147:5340–5351.
    1. Manco M. Endotoxin as a missed link among all the metabolic abnormalities in the metabolic syndrome. Atherosclerosis. 2009;206:36; author reply–37.
    1. Stoll L. L, Denning G. M, Weintraub N. L. Potential role of endotoxin as a proinflammatory mediator of atherosclerosis. Arterioscler Thromb Vasc Biol. 2004;24:2227–2236.
    1. Spor A, Koren O, Ley R. Unravelling the effects of the environment and host genotype on the gut microbiome. Nat Rev Microbiol. 9:279–290.
    1. Kien C. L, Schmitz-Brown M, Solley T, Sun D, Frankel W. L. Increased colonic luminal synthesis of butyric acid is associated with lowered colonic cell proliferation in piglets. J Nutr. 2006;136:64–69.
    1. Turnbaugh P. J, Ley R. E, Mahowald M. A, Magrini V, Mardis E. R, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–1031.
    1. Macdonald T. T, Monteleone G. Immunity, inflammation, and allergy in the gut. Science. 2005;307:1920–1925.
    1. Bettelheim K. A, Breadon A, Faiers M. C, O'Farrell S. M, Shooter R. A. The origin of O serotypes of Escherichia coli in babies after normal delivery. J Hyg (Lond) 1974;72:67–70.
    1. Bezirtzoglou E. The intestinal microflora during the first weeks of life. Anaerobe. 1997;3:173–177.
    1. Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. 2003;21:335–376.
    1. Beutler B. Inferences, questions and possibilities in Toll-like receptor signalling. Nature. 2004;430:257–263.
    1. Fogelstrand L, Hulthe J, Hulten L. M, Wiklund O, Fagerberg B. Monocytic expression of CD14 and CD18, circulating adhesion molecules and inflammatory markers in women with diabetes mellitus and impaired glucose tolerance. Diabetologia. 2004;47:1948–1952.
    1. Tsukumo D. M, Carvalho-Filho M. A, Carvalheira J. B, Prada P. O, Hirabara S. M, et al. Loss-of-function mutation in Toll-like receptor 4 prevents diet-induced obesity and insulin resistance. Diabetes. 2007;56:1986–1998.
    1. Shi H, Kokoeva M. V, Inouye K, Tzameli I, Yin H, et al. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest. 2006;116:3015–3025.
    1. Kuo L. H, Tsai P. J, Jiang M. J, Chuang Y. L, Yu L, et al. Toll-like receptor 2 deficiency improves insulin sensitivity and hepatic insulin signalling in the mouse. Diabetologia. 54:168–179.
    1. Ehses J. A, Meier D. T, Wueest S, Rytka J, Boller S, et al. Toll-like receptor 2-deficient mice are protected from insulin resistance and beta cell dysfunction induced by a high-fat diet. Diabetologia. 53:1795–1806.
    1. Vijay-Kumar M, Aitken J. D, Carvalho F. A, Cullender T. C, Mwangi S, et al. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science. 328:228–231.
    1. Cani P. D, Possemiers S, Van de Wiele T, Guiot Y, Everard A, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut. 2009;58:1091–1103.
    1. Ozcan U, Cao Q, Yilmaz E, Lee A. H, Iwakoshi N. N, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004;306:457–461.
    1. Ozcan L, Ergin A. S, Lu A, Chung J, Sarkar S, et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab. 2009;9:35–51.
    1. Sutmuller R. P, den Brok M. H, Kramer M, Bennink E. J, Toonen L. W, et al. Toll-like receptor 2 controls expansion and function of regulatory T cells. J Clin Invest. 2006;116:485–494.
    1. Barnard R. J, Roberts C. K, Varon S. M, Berger J. J. Diet-induced insulin resistance precedes other aspects of the metabolic syndrome. J Appl Physiol. 1998;84:1311–1315.
    1. Barnard R. J, Faria D. J, Menges J. E, Martin D. A. Effects of a high-fat, sucrose diet on serum insulin and related atherosclerotic risk factors in rats. Atherosclerosis. 1993;100:229–236.
    1. Himes R. W, Smith C. W. Tlr2 is critical for diet-induced metabolic syndrome in a murine model. FASEB J. 24:731–739.
    1. Backhed F, Ding H, Wang T, Hooper L. V, Koh G. Y, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A. 2004;101:15718–15723.
    1. Rabot S, Membrez M, Bruneau A, Gerard P, Harach T, et al. Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J. 24:4948–4959.
    1. Kellermayer R, Dowd S. E, Harris R. A, Balasa A, Schaible T. D, et al. Colonic mucosal DNA methylation, immune response, and microbiome patterns in Toll-like receptor 2-knockout mice. FASEB J. 25:1449–1460.
    1. Ley R. E, Turnbaugh P. J, Klein S, Gordon J. I. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–1023.
    1. Ley R. E, Backhed F, Turnbaugh P, Lozupone C. A, Knight R. D, et al. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102:11070–11075.
    1. Backhed F, Manchester J. K, Semenkovich C. F, Gordon J. I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A. 2007;104:979–984.
    1. Manco M, Putignani L, Bottazzo G. F. Gut microbiota, lipopolysaccharides, and innate immunity in the pathogenesis of obesity and cardiovascular risk. Endocr Rev. 31:817–844.
    1. Purohit V, Bode J. C, Bode C, Brenner D. A, Choudhry M. A, et al. Alcohol, intestinal bacterial growth, intestinal permeability to endotoxin, and medical consequences: summary of a symposium. Alcohol. 2008;42:349–361.
    1. Wigg A. J, Roberts-Thomson I. C, Dymock R. B, McCarthy P. J, Grose R. H, et al. The role of small intestinal bacterial overgrowth, intestinal permeability, endotoxaemia, and tumour necrosis factor alpha in the pathogenesis of non-alcoholic steatohepatitis. Gut. 2001;48:206–211.
    1. Miele L, Valenza V, La Torre G, Montalto M, Cammarota G, et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology. 2009;49:1877–1887.
    1. Adlerberth I, Wold A. E. Establishment of the gut microbiota in Western infants. Acta Paediatr. 2009;98:229–238.
    1. Lichtman S. N, Keku J, Schwab J. H, Sartor R. B. Hepatic injury associated with small bowel bacterial overgrowth in rats is prevented by metronidazole and tetracycline. Gastroenterology. 1991;100:513–519.
    1. Wang Z, Xiao G, Yao Y, Guo S, Lu K, et al. The role of bifidobacteria in gut barrier function after thermal injury in rats. J Trauma. 2006;61:650–657.
    1. Griffiths E. A, Duffy L. C, Schanbacher F. L, Qiao H, Dryja D, et al. In vivo effects of bifidobacteria and lactoferrin on gut endotoxin concentration and mucosal immunity in Balb/c mice. Dig Dis Sci. 2004;49:579–589.
    1. Cario E, Gerken G, Podolsky D. K. Toll-like receptor 2 controls mucosal inflammation by regulating epithelial barrier function. Gastroenterology. 2007;132:1359–1374.
    1. Cario E. Bacterial interactions with cells of the intestinal mucosa: Toll-like receptors and NOD2. Gut. 2005;54:1182–1193.
    1. Kellermayer R, Dowd S. E, Harris R. A, Balasa A, Schaible T. D, et al. Colonic mucosal DNA methylation, immune response, and microbiome patterns in Toll-like receptor 2-knockout mice. FASEB J
    1. Hotamisligil G. S, Peraldi P, Budavari A, Ellis R, White M. F, et al. IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science. 1996;271:665–668.
    1. Song M. J, Kim K. H, Yoon J. M, Kim J. B. Activation of Toll-like receptor 4 is associated with insulin resistance in adipocytes. Biochem Biophys Res Commun. 2006;346:739–745.
    1. Laflamme N, Echchannaoui H, Landmann R, Rivest S. Cooperation between toll-like receptor 2 and 4 in the brain of mice challenged with cell wall components derived from gram-negative and gram-positive bacteria. Eur J Immunol. 2003;33:1127–1138.
    1. Tanti J. F, Gremeaux T, van Obberghen E, Le Marchand-Brustel Y. Serine/threonine phosphorylation of insulin receptor substrate 1 modulates insulin receptor signaling. J Biol Chem. 1994;269:6051–6057.
    1. Saltiel A. R, Kahn C. R. Insulin signalling and the regulation of glucose and lipid metabolism. Nature. 2001;414:799–806.
    1. Zheng Y, Rudensky A. Y. Foxp3 in control of the regulatory T cell lineage. Nat Immunol. 2007;8:457–462.
    1. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133:775–787.
    1. Maloy K. J, Salaun L, Cahill R, Dougan G, Saunders N. J, et al. CD4+CD25+ T(R) cells suppress innate immune pathology through cytokine-dependent mechanisms. J Exp Med. 2003;197:111–119.
    1. Murphy T. J, Ni Choileain N, Zang Y, Mannick J. A, Lederer J. A. CD4+CD25+ regulatory T cells control innate immune reactivity after injury. J Immunol. 2005;174:2957–2963.
    1. Nguyen L. T, Jacobs J, Mathis D, Benoist C. Where FoxP3-dependent regulatory T cells impinge on the development of inflammatory arthritis. Arthritis Rheum. 2007;56:509–520.
    1. Takeuchi O, Hoshino K, Kawai T, Sanjo H, Takada H, et al. Differential roles of TLR2 and TLR4 in recognition of gram-negative and gram-positive bacterial cell wall components. Immunity. 1999;11:443–451.
    1. Campos M. A, Rosinha G. M, Almeida I. C, Salgueiro X. S, Jarvis B. W, et al. Role of Toll-like receptor 4 in induction of cell-mediated immunity and resistance to Brucella abortus infection in mice. Infect Immun. 2004;72:176–186.
    1. Mombaerts P, Iacomini J, Johnson R. S, Herrup K, Tonegawa S, et al. RAG-1-deficient mice have no mature B and T lymphocytes. Cell. 1992;68:869–877.
    1. Araujo E. P, De Souza C. T, Ueno M, Cintra D. E, Bertolo M. B, et al. Infliximab restores glucose homeostasis in an animal model of diet-induced obesity and diabetes. Endocrinology. 2007;148:5991–5997.
    1. DeFronzo R. A, Tobin J. D, Andres R. Glucose clamp technique: a method for quantifying insulin secretion and resistance. Am J Physiol. 1979;237:E214–E223.
    1. Hirabara S. M, Silveira L. R, Alberici L. C, Leandro C. V, Lambertucci R. H, et al. Acute effect of fatty acids on metabolism and mitochondrial coupling in skeletal muscle. Biochim Biophys Acta. 2006;1757:57–66.
    1. Chen W. Y, Bailey E. C, McCune S. L, Dong J. Y, Townes T. M. Reactivation of silenced, virally transduced genes by inhibitors of histone deacetylase. Proc Natl Acad Sci U S A. 1997;94:5798–5803.
    1. Ramirez-Alcantara V, LoGuidice A, Boelsterli U. A. Protection from diclofenac-induced small intestinal injury by the JNK inhibitor SP600125 in a mouse model of NSAID-associated enteropathy. Am J Physiol Gastrointest Liver Physiol. 2009;297:G990–G998.
    1. Takashima K, Matsunaga N, Yoshimatsu M, Hazeki K, Kaisho T, et al. Analysis of binding site for the novel small-molecule TLR4 signal transduction inhibitor TAK-242 and its therapeutic effect on mouse sepsis model. Br J Pharmacol. 2009;157:1250–1262.
    1. Schumann A, Nutten S, Donnicola D, Comelli E. M, Mansourian R, et al. Neonatal antibiotic treatment alters gastrointestinal tract developmental gene expression and intestinal barrier transcriptome. Physiol Genomics. 2005;23:235–245.
    1. Membrez M, Blancher F, Jaquet M, Bibiloni R, Cani P. D, et al. Gut microbiota modulation with norfloxacin and ampicillin enhances glucose tolerance in mice. FASEB J. 2008;22:2416–2426.
    1. Meyer F, Paarmann D, D'Souza M, Olson R, Glass E. M, et al. The metagenomics RAST server - a public resource for the automatic phylogenetic and functional analysis of metagenomes. BMC Bioinformatics. 2008;9:386.

Source: PubMed

3
Předplatit