Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Sci Adv ; 9(35): eadg1129, 2023 09.
Article in English | MEDLINE | ID: mdl-37656793

ABSTRACT

Although the etiology of rheumatoid arthritis (RA) is unknown, a strong genetic predisposition and the presence of preclinical antibodies before the onset of symptoms is documented. An expansion of Eggerthella lenta is associated with severe disease in RA. Here, using a humanized mouse model of collagen-induced arthritis, we determined the impact of E. lenta abundance on RA severity. Naïve mice gavaged with E. lenta produce preclinical rheumatoid factor and, when induced for arthritis, develop severe disease. The augmented antibody response was much higher in female mice, and among patients with RA, women had higher average load of E. lenta. Expansion of E. lenta increased CXCL5 and CD4 T cells, and both interleukin-17- and interferon-γ-producing B cells. Further, E. lenta gavage caused gut dysbiosis and decline in amino acids and nicotinamide adenine dinucleotide with an increase in microbe-dependent bile acids and succinyl carnitine causing systemic senescent-like inflammation.


Subject(s)
Actinobacteria , Arthritis, Rheumatoid , Female , Animals , Mice , Arthritis, Rheumatoid/etiology , Inflammation , Autoantibodies
2.
Gastroenterology ; 164(2): 256-271.e10, 2023 02.
Article in English | MEDLINE | ID: mdl-36272457

ABSTRACT

BACKGROUND & AIMS: Although T-cell intrinsic expression of G9a has been associated with murine intestinal inflammation, mechanistic insight into the role of this methyltransferase in human T-cell differentiation is ill defined, and manipulation of G9a function for therapeutic use against inflammatory disorders is unexplored. METHODS: Human naive T cells were isolated from peripheral blood and differentiated in vitro in the presence of a G9a inhibitor (UNC0642) before being characterized via the transcriptome (RNA sequencing), chromatin accessibility (assay for transposase-accessible chromatin by sequencing), protein expression (cytometry by time of flight, flow cytometry), metabolism (mitochondrial stress test, ultrahigh performance liquid chromatography-tandem mas spectroscopy) and function (T-cell suppression assay). The in vivo role of G9a was assessed using 3 murine models. RESULTS: We discovered that pharmacologic inhibition of G9a enzymatic function in human CD4 T cells led to spontaneous generation of FOXP3+ T cells (G9a-inibitors-T regulatory cells [Tregs]) in vitro that faithfully reproduce human Tregs, functionally and phenotypically. Mechanistically, G9a inhibition altered the transcriptional regulation of genes involved in lipid biosynthesis in T cells, resulting in increased intracellular cholesterol. Metabolomic profiling of G9a-inibitors-Tregs confirmed elevated lipid pathways that support Treg development through oxidative phosphorylation and enhanced lipid membrane composition. Pharmacologic G9a inhibition promoted Treg expansion in vivo upon antigen (gliadin) stimulation and ameliorated acute trinitrobenzene sulfonic acid-induced colitis secondary to tissue-specific Treg development. Finally, Tregs lacking G9a expression (G9a-knockout Tregs) remain functional chronically and can rescue T-cell transfer-induced colitis. CONCLUSION: G9a inhibition promotes cholesterol metabolism in T cells, favoring a metabolic profile that facilitates Treg development in vitro and in vivo. Our data support the potential use of G9a inhibitors in the treatment of immune-mediated conditions including inflammatory bowel disease.


Subject(s)
CD4-Positive T-Lymphocytes , Colitis , Mice , Humans , Animals , Lipid Metabolism , T-Lymphocytes, Regulatory/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/genetics , Chromatin , Inflammation , Cholesterol , Lipids , Forkhead Transcription Factors/metabolism
3.
BMC Microbiol ; 22(1): 8, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34983374

ABSTRACT

BACKGROUND: Type 1 diabetes (T1D) is an autoimmune disease that is increasing in prevalence worldwide. One of the contributing factors to the pathogenesis of T1D is the composition of the intestinal microbiota, as has been demonstrated. in T1D patients, with some studies demonstrating a deficiency in their levels of Prevotella. We have isolated a strain of Prevotella histicola from a duodenal biopsy that has anti-inflammatory properties, and in addition, alters the development of autoimmune diseases in mouse models. Therefore, our hypothesis is that the oral administration of P. histicola might delay the development of T1D in the non-obese diabetic (NOD) mice. To assess this, we used the following materials and methods. Female NOD mice (ages 5-8 weeks) were administered every other day P. histicola that was cultured in-house. Blood glucose levels were measured every other week. Mice were sacrificed at various time points for histopathological analysis of the pancreas. Modulation of immune response by the commensal was tested by analyzing regulatory T-cells and NKp46+ cells using flow cytometry and intestinal cytokine mRNA transcript levels using quantitative RT-PCR. For microbial composition, 16 s rRNA gene analysis was conducted on stool samples collected at various time points. RESULTS: Administration of P. histicola in NOD mice delayed the onset of T1D. Beta diversity in the fecal microbiomes demonstrated that the microbial composition of the mice administered P. histicola was different from those that were not treated. Treatment with P. histicola led to a significant increase in regulatory T cells with a concomitant decrease in NKp46+ cells in the pancreatic lymph nodes as compared to the untreated group after 5 weeks of treatment. CONCLUSIONS: These observations suggest that P. histicola treatment delayed onset of diabetes by increasing the levels of regulatory T cells in the pancreatic lymph nodes. This preliminary work supports the rationale that enteral exposure to a non pathogenic commensal P. histicola be tested as a future therapy for T1D.


Subject(s)
Diabetes Mellitus, Type 1/diet therapy , Gastrointestinal Microbiome/physiology , Prevotella/physiology , Probiotics/administration & dosage , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Cytokines/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Duodenum/immunology , Duodenum/microbiology , Feces/microbiology , Female , Humans , Mice , Mice, Inbred NOD , Pancreas/immunology , Pancreas/pathology
4.
Clin Immunol ; 230: 108813, 2021 09.
Article in English | MEDLINE | ID: mdl-34333094

ABSTRACT

Cigarette smoking has been implicated in the pathogenesis of seropositive rheumatoid arthritis (RA), as well as RA-associated lung disease. Fibrotic interstitial lung disease as well as emphysema occur in RA and cause substantial morbidity. We used arthritis-susceptible HLA-DQ8 transgenic mice to generate RA-associated lung disease. Mice were exposed to cigarette smoke (CS) prior to induction of arthritis, and subsequently injected with a low dose of bleomycin intra-tracheally to induce lung injury. Exposure of arthritic mice to both CS and bleomycin led to a significant reduction in lung compliance consistent with development of diffuse lung disease. Morphologic evaluation of the lung demonstrated areas of emphysematous change and co-existent fibrosis, consistent with a combined pattern of fibrosis and emphysema. These changes were accompanied by inflammatory cell infiltration and upregulation of fibrosis-associated genes. This humanized mouse model can serve as a valuable research tool to understand the pathogenesis of RA associated lung disease.


Subject(s)
Arthritis, Rheumatoid/complications , Lung Diseases, Interstitial/etiology , Animals , Arthritis, Rheumatoid/etiology , Bleomycin/toxicity , Cigarette Smoking/adverse effects , Collagen/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Lung/metabolism , Lung/pathology , Lung Compliance/drug effects , Lung Diseases, Interstitial/pathology , Male , Mice , Mice, Transgenic , Pulmonary Emphysema/etiology , Pulmonary Fibrosis/etiology
5.
Front Immunol ; 12: 609644, 2021.
Article in English | MEDLINE | ID: mdl-34017324

ABSTRACT

Bacterial therapeutics are the emergent alternatives in treating autoimmune diseases such as Rheumatoid Arthritis [RA]. P. histicola MCI 001 is one such therapeutic bacterium that has been proven to treat autoimmune diseases such as RA and multiple sclerosis [MS] in animal models. The present study characterized P. histicola MCI 001 isolated from a human duodenal biopsy, and evaluated its impact on the gut microbial and metabolic profile in a longitudinal study using the collagen-induced arthritis model in HLA-DQ8.AEo transgenic mice. P. histicola MCI 001 though closely related to the type strain of P. histicola, DSM 19854, differed in utilizing glycerol. In culture, P. histicola MCI 001 produced vitamins such as biotin and folate, and was involved in digesting complex carbohydrates and production of acetate. Colonization study showed that duodenum was the predominant niche for the gavaged MCI 001. A longitudinal follow-up of gut microbial profile in arthritic mice treated with MCI 001 suggested that dysbiosis caused due to arthritis was partially restored to the profile of naïve mice after treatment. A taxon-level analysis suggested an expansion of intestinal genus Allobaculum in MCI001 treated arthritic mice. Eubiosis achieved post treatment with P. histicola MCI 001 was also reflected in the increased production of short-chain fatty acids [SCFAs]. Present study suggests that the treatment with P. histicola MCI 001 leads to an expansion of Allobaculum by increasing the availability of simple carbohydrates and acetate. Restoration of microbial profile and metabolites like butyrate induce immune and gut homeostasis.


Subject(s)
Biological Therapy/methods , Butyrates/metabolism , Prevotella/physiology , Symbiosis , Adaptation, Physiological , Animals , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/therapy , Bile Acids and Salts/pharmacology , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Gastric Juice , Gastrointestinal Microbiome , Humans , Hydrogen-Ion Concentration , Mice , Mice, Transgenic , Prevotella/classification , Prevotella/drug effects , Prevotella/genetics
6.
Anaerobe ; 68: 102237, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32721554

ABSTRACT

The human intestinal tract is colonized by a large number of diverse microorganisms that play various important physiologic functions. In inflammatory gut diseases including celiac disease (CeD), a dysbiotic state of microbiome has been observed. Interestingly, this perturbed microbiome is normalized towards eubiosis in patients showing recovery after treatment. The treatment has been observed to increase the abundance of beneficial microbes in comparison to non-treated patients. In this study, we investigated the effect of Prevotella histicola or Prevotella melaninogenica, isolated from the duodenum of a treated CeD patient, on the induction and maintenance of oral tolerance to gliadin, a CeD associated subgroup of gluten proteins, in NOD.DQ8.ABo transgenic mice. Conventionally raised mice on a gluten free diet were orally gavaged with bacteria before and after injection with pepsin trypsin digested gliadin (PTD-gliadin). P. histicola suppressed the cellular response to gliadin, whereas P. melaninogenica failed to suppress an immune response against gliadin. Interestingly, tolerance to gliadin in NOD.DQ8.ABo mice may be associated with gut microbiota as mice gavaged with P melaninogenica harbored a different microbial diversity as compared to P. histicola treated mice. This study provides experimental evidence that gut microbes like P. histicola from treated patients can suppress the immune response against gliadin epitopes.


Subject(s)
Celiac Disease/immunology , Celiac Disease/microbiology , Gastrointestinal Microbiome , Gliadin/immunology , T-Lymphocytes/immunology , Animals , Female , Humans , Immune Tolerance , Male , Mice , Mice, Inbred NOD , Prevotella/immunology , Prevotella/physiology , Prevotella melaninogenica/immunology , Prevotella melaninogenica/physiology
7.
Sci Rep ; 9(1): 19061, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31836763

ABSTRACT

Major histocompatibility complex II (MHC II) molecules are involved in antigen presentation and the development of a functional adaptive immune response. Evolutionary selection for MHC molecules that effectively clear infectious agents provides an advantage to humans. However, certain class II molecules are associated with autoimmune diseases. In this study we infected autoimmune-susceptible DRB1*0401.AEo and non-susceptible *0402.AEo mice with H1N1 influenza and determined clearance and protective immunity to H3N2 virus. *0401 mice generated a robust TLR-triggered immune response and cleared H1N1 influenza virus infection. After vaccination and challenge with H1N1, *0401 mice, when challenged with H3N2, generated cross-protective immunity to heterosubtypic H3N2 influenza strain whereas *0402 mice cleared the H1N1 infection but did not generate cross-protective immunity against the H3N2 influenza strain. The intracellular trafficking route of MHCII revealed that *0401 molecules traffic through the late endosome/lysosomes while *0402 molecules traffic into early endosomes. This suggested that trafficking of MHCII could affect the functional output of the innate immune response and clearance of viral infections. Also, DRB1*0401 mice live longer than HLA-DRB1*0402 mice. The study provides a potential hypothesis for evolutionary selection of *0401 molecule, even though it is associated with autoreactivity, which may be dependent on the availability of peptide repertoire of self-antigens.


Subject(s)
Histocompatibility Antigens Class II/genetics , Immunity/genetics , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Polymorphism, Genetic , Aging/immunology , Amino Acid Sequence , Animals , Cross Reactions/genetics , Endosomes/metabolism , Histocompatibility Antigens Class II/chemistry , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Lysosomes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Survival Analysis , T-Lymphocytes/immunology , Vaccination
8.
Mil Med ; 184(Suppl 1): 529-536, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30901468

ABSTRACT

OBJECTIVE: Although the etiology of rheumatoid arthritis (RA) is unknown, recent studies have led to the concept that gut dysbiosis may be involved in onset. In this study, we aimed to determine if human gut commensals modulate the immune response and gut epithelial integrity in DQ8 mice. METHODS: DQ8 mice were orally gavaged with RA-associated (Eggerthella lenta or Collinsella aerofaciens) and non-associated (Prevotella histicola or Bifidobacterium sp.) on alternate days for 1 week in naïve mice. Some mice were immunized with type II collagen and oral gavage continued for 6 weeks and followed for arthritis. Epithelial integrity was done by FITC-Dextran assay. In addition, cytokines were measured in sera by ELISA and various immune cells were quantified using flow cytometry. RESULTS: Gut permeability was increased by the RA-associated bacteria and was sex and age-dependent. In vivo and in vitro observations showed that the RA-non-associated bacteria outgrow the RA-associated bacteria when gavaged or cultured together. Mice gavaged with the RA-non-associated bacteria produced lower levels of pro-inflammatory MCP-1 and MCP-3 and had lower numbers of Inflammatory monocytes CD11c+Ly6c+, when compared to controls. E. lenta treated naïve mice produce Th17 cytokines. CONCLUSIONS: Our studies suggest that gut commensals influence immune response in and away from the gut by changing the gut permeability and immunity. Dysbiosis helps the growth of RA-associated bacteria and reduces the beneficial bacteria.


Subject(s)
Autoimmunity/physiology , Gastrointestinal Microbiome/physiology , Permeability , Animals , Bifidobacterium/pathogenicity , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Mice , Prevalence , Symbiosis/physiology
9.
Cell Rep ; 20(6): 1269-1277, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28793252

ABSTRACT

The human gut is colonized by a large number of microorganisms (∼1013 bacteria) that support various physiologic functions. A perturbation in the healthy gut microbiome might lead to the development of inflammatory diseases, such as multiple sclerosis (MS). Therefore, gut commensals might provide promising therapeutic options for treating MS and other diseases. We report the identification of human gut-derived commensal bacteria, Prevotella histicola, which can suppress experimental autoimmune encephalomyelitis (EAE) in a human leukocyte antigen (HLA) class II transgenic mouse model. P. histicola suppresses disease through the modulation of systemic immune responses. P. histicola challenge led to a decrease in pro-inflammatory Th1 and Th17 cells and an increase in the frequencies of CD4+FoxP3+ regulatory T cells, tolerogenic dendritic cells, and suppressive macrophages. Our study provides evidence that the administration of gut commensals may regulate a systemic immune response and may, therefore, have a possible role in treatment strategies for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Gastrointestinal Microbiome , Prevotella/immunology , Probiotics/therapeutic use , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/microbiology , Forkhead Transcription Factors/metabolism , Genes, MHC Class II , Humans , Macrophages/immunology , Mice , Prevotella/pathogenicity , Th1 Cells/immunology , Th17 Cells/immunology
10.
Anaerobe ; 48: 37-41, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28684342

ABSTRACT

We designed species-specific primers and developed a qPCR method for enumerating P. histicola from intestinal samples. The two designed primer sets showed specificity for the target 16S rRNA gene of P. histicola. The absolute qPCR method was sensitive to quantify as few as 103 colony-forming units (CFU) in the gut.


Subject(s)
Bacteroidaceae Infections/diagnosis , Intestines/microbiology , Prevotella/isolation & purification , Real-Time Polymerase Chain Reaction/methods , Animals , Bacteroidaceae Infections/microbiology , DNA Primers/genetics , DNA, Bacterial/genetics , Mice , Mice, Transgenic , RNA, Ribosomal, 16S/genetics
11.
Clin Immunol ; 178: 10-19, 2017 05.
Article in English | MEDLINE | ID: mdl-26057130

ABSTRACT

Rheumatoid arthritis (RA) occurs two times more often in women than men. B cell depletion has been shown to be efficacious in treating RA. Our previous studies suggested that antigen presentation via B cells results in a sex-specific immune response in DR4 and DR4/DQ8 mice. Here we evaluated the mechanism of efficacy of the B cell depletion in treating arthritis-susceptible DQ8 mice. The data show that arthritic DQ8 mice treated with anti-CD20 antibody in therapeutic protocols show milder disease severity in females as compared to males, which is associated with decreased antibodies to citrullinated proteins and reduced levels of IL-23 and CCL5. Treatment led to significantly increased numbers of T regulatory and monocyte-derived suppressor F4/80+Gr1hi cells in females as compared to male DQ8 mice. Our observations suggest that therapeutic strategies that target B cells may benefit females while functions of DCs might be relatively more important for men than women.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , B-Lymphocytes/immunology , Chemokine CCL5/immunology , Interleukin-23/immunology , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antirheumatic Agents/pharmacology , Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , B-Lymphocytes/drug effects , CD28 Antigens/immunology , CD40 Antigens/immunology , Cell Proliferation , Chemokine CCL5/drug effects , Chemokines/drug effects , Chemokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Flow Cytometry , HLA-DQ beta-Chains/genetics , Humans , Interleukin-23/drug effects , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Rituximab/pharmacology , Sex Characteristics , T-Lymphocytes, Regulatory/drug effects
12.
PLoS One ; 11(9): e0162341, 2016.
Article in English | MEDLINE | ID: mdl-27602574

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease marked by chronic synovial inflammation and both, genetic and environmental factors are involved in its pathogenesis. Human leukocyte antigen (HLA) DRB1*0401 is associated with susceptibility to develop RA, while cigarette smoke (CS) exposure promotes seropositive disease with increased severity in DRB1*0401+ individuals. Smokers have higher levels of antibodies against citrullinated peptides. In this study, we determined whether the response to a known autoantigen, Vimentin (Vim) is shared epitope specific and how CS influences this response using transgenic-mice carrying RA-susceptible,*0401, and -resistant, *0402, genes. Following relatively brief exposure to CS, peptidyl arginine deiminase (PAD) enzyme expression was increased in murine lungs. Cigarette smoking led to production of Interferon (IFN)-γ with reduced levels of Interleukin (IL)-10 by splenocytes of *0401 mice. In contrast, CS augmented Th2 cytokines along with T-regulatory cells in *0402 mice. An increase in levels of antibodies to native and citrullinated Vim was observed in naïve mice of both strains following CS exposure. Our data showed that both arthritis-susceptible and -resistant mice can generate cellular and humoral immunity to Vim; however CS-induced modulation of host immunity is dependent on the interaction with the host HLA genes.


Subject(s)
Arthritis, Rheumatoid/immunology , Disease Resistance/immunology , Smoking/adverse effects , Vimentin/immunology , Animals , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/genetics , Cell Proliferation , Cytokines/metabolism , Disease Resistance/genetics , Disease Susceptibility , Epitopes/immunology , Female , HLA-DRB1 Chains/genetics , Humans , Hydrolases/metabolism , Immunity, Humoral , Lung/enzymology , Lung/pathology , Lymphocyte Count , Mice, Inbred C57BL , Mice, Transgenic , Protein-Arginine Deiminases , T-Lymphocytes/immunology
13.
Sci Rep ; 6: 28484, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27346372

ABSTRACT

Multiple sclerosis (MS) is an immune-mediated disease, the etiology of which involves both genetic and environmental factors. The exact nature of the environmental factors responsible for predisposition to MS remains elusive; however, it's hypothesized that gastrointestinal microbiota might play an important role in pathogenesis of MS. Therefore, this study was designed to investigate whether gut microbiota are altered in MS by comparing the fecal microbiota in relapsing remitting MS (RRMS) (n = 31) patients to that of age- and gender-matched healthy controls (n = 36). Phylotype profiles of the gut microbial populations were generated using hypervariable tag sequencing of the V3-V5 region of the 16S ribosomal RNA gene. Detailed fecal microbiome analyses revealed that MS patients had distinct microbial community profile compared to healthy controls. We observed an increased abundance of Psuedomonas, Mycoplana, Haemophilus, Blautia, and Dorea genera in MS patients, whereas control group showed increased abundance of Parabacteroides, Adlercreutzia and Prevotella genera. Thus our study is consistent with the hypothesis that MS patients have gut microbial dysbiosis and further study is needed to better understand their role in the etiopathogenesis of MS.


Subject(s)
Gastrointestinal Microbiome/genetics , Gastrointestinal Tract/microbiology , Multiple Sclerosis/microbiology , Adult , Dysbiosis/microbiology , Female , Humans , Male , Microbiota/genetics , RNA, Ribosomal, 16S/genetics
14.
Arthritis Rheumatol ; 68(12): 2878-2888, 2016 12.
Article in English | MEDLINE | ID: mdl-27337150

ABSTRACT

OBJECTIVE: The gut microbiome regulates host immune homeostasis. Rheumatoid arthritis (RA) is associated with intestinal dysbiosis. This study was undertaken to test the ability of a human gut-derived commensal to modulate immune response and treat arthritis in a humanized mouse model. METHODS: We isolated a commensal bacterium, Prevotella histicola, that is native to the human gut and has systemic immune effects when administered enterally. Arthritis-susceptible HLA-DQ8 mice were immunized with type II collagen and treated with P histicola. Disease incidence, onset, and severity were monitored. Changes in gut epithelial proteins and immune response as well as systemic cellular and humoral immune responses were studied in treated mice. RESULTS: When treated with P histicola in prophylactic or therapeutic protocols, DQ8 mice exhibited significantly decreased incidence and severity of arthritis compared to controls. The microbial mucosal modulation of arthritis was dependent on regulation by CD103+ dendritic cells and myeloid suppressors (CD11b+Gr-1+ cells) and by generation of Treg cells (CD4+CD25+FoxP3+) in the gut, resulting in suppression of antigen-specific Th17 responses and increased transcription of interleukin-10. Treatment with P histicola led to reduced intestinal permeability by increasing expression of enzymes that produce antimicrobial peptides as well as tight junction proteins (zonula occludens 1 and occludin). However, the innate immune response via Toll-like receptor 4 (TLR-4) and TLR-9 was not affected in treated mice. CONCLUSION: Our results demonstrate that enteral exposure to P histicola suppresses arthritis via mucosal regulation. P histicola is a unique commensal that can be explored as a novel therapy for RA and may have few or no side effects.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Dendritic Cells/immunology , Gastrointestinal Microbiome/immunology , Intestines/immunology , Prevotella/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , Cell Proliferation , Chemokines/genetics , Chemokines/immunology , Cytokines/genetics , Cytokines/immunology , Flow Cytometry , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/microbiology , Mice , Mice, Transgenic , Occludin/metabolism , Permeability , Prevotella melaninogenica/immunology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Th17 Cells/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 9/immunology , Zonula Occludens-1 Protein/metabolism
15.
Clin Immunol ; 159(2): 154-62, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25956531

ABSTRACT

Autoimmune diseases like rheumatoid arthritis are multifactorial in nature, requiring both genetic and environmental factors for onset. Increased predisposition of females to a wide range of autoimmune diseases points to a gender bias in the multifactorial etiology of these disorders. However, the existing evidence to date has not provided any conclusive mechanism of gender-bias beyond the role of hormones and sex chromosomes. The gut microbiome, which impacts the innate and adaptive branches of immunity, not only influences the development of autoimmune disorders but may interact with sex-hormones to modulate disease progression and sex-bias. Here, we review the current information on gender bias in autoimmunity and discuss the potential of microbiome-derived biomarkers to help unravel the complex interplay between genes, environment and hormones in rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid/immunology , Autoimmunity/immunology , Gastrointestinal Microbiome/immunology , Gonadal Steroid Hormones/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/microbiology , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/microbiology , Female , Gene-Environment Interaction , Gonadal Steroid Hormones/metabolism , Humans , Male , Sex Factors
16.
J Immunol ; 193(10): 4859-70, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25339670

ABSTRACT

Multiple sclerosis is an inflammatory, demyelinating disease of the CNS of presumed autoimmune origin. Of all the genetic factors linked with multiple sclerosis, MHC class II molecules have the strongest association. Generation of HLA class II transgenic (Tg) mice has helped to elucidate the role of HLA class II genes in chronic inflammatory and demyelinating diseases. We have shown that the human HLA-DRB1*0301 gene predisposes to proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE), whereas HLA-DQß1*0601 (DQ6) was resistant. We also showed that the DQ6 molecule protects from EAE in DRB1*0301.DQ6 double-Tg mice by producing anti-inflammatory IFN-γ. HLA-DQß1*0302 (DQ8) Tg mice were also resistant to PLP(91-110)-induced EAE, but production of proinflammatory IL-17 exacerbated disease in DRB1*0301.DQ8 mice. To further confirm the role of IFN-γ in protection, we generated DRB1*0301.DQ8 mice lacking IFN-γ (DRB1*0301.DQ8.IFN-γ(-/-)). Immunization with PLP(91-110) peptide caused atypical EAE in DRB1*0301.DQ8.IFN-γ(-/-) mice characterized by ataxia, spasticity, and dystonia, hallmarks of brain-specific disease. Severe brain-specific inflammation and demyelination in DRB1*0301.DQ8.IFN-γ(-/-) mice with minimal spinal cord pathology further confirmed brain-specific pathology. Atypical EAE in DRB1*0301.DQ8.IFN-γ(-/-) mice was associated with increased encephalitogenicity of CD4 T cells and their ability to produce greater levels of IL-17 and GM-CSF compared with DRB1*0301.DQ8 mice. Further, areas with demyelination showed increased presence of CD68(+) inflammatory cells, suggesting an important role for monocytes/microglia in causing brain pathology. Thus, our study supports a protective role for IFN-γ in the demyelination of brain through downregulation of IL-17/GM-CSF and induction of neuroprotective factors in the brain by monocytes/microglial cells.


Subject(s)
Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interferon-gamma/immunology , Interleukin-17/immunology , Microglia/immunology , Monocytes/immunology , Animals , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , HLA-DQ Antigens/genetics , HLA-DQ Antigens/immunology , HLA-DQ beta-Chains/genetics , HLA-DQ beta-Chains/immunology , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/immunology , Humans , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interleukin-17/genetics , Mice , Mice, Knockout , Microglia/pathology , Monocytes/pathology , Myelin Proteolipid Protein , Signal Transduction
17.
Eur J Immunol ; 44(11): 3429-38, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25103892

ABSTRACT

HLA-DRB1*0401 expression in humans has been associated with a predisposition to developing rheumatoid arthritis (RA) and collagen-induced arthritis (CIA), while HLA-DRB1*0402 is not associated with susceptibility. Here, we determined if mice transgenic (Tg) for human *0401 have a CD4+ T-cell repertoire that predetermines proinflammatory cytokine production. The data show that both *0401 and *0402 Tg mice can produce TH1/TH17 cytokines, although the kinetics of response may be different. However, in the context of antigen-specific responses in a CIA model, *0402 Tg mice generate a TH2 response that may explain their resistance to developing arthritis. In addition, a significant subset of naïve CD4+ T cells from *0402 Tg mice can be activated in polarizing conditions to differentiate into Treg cells that produce IFN-γ. *0401 Tg mice harbor memory CD4+ T cells that differentiate into IL-17(+) cells in various polarizing conditions. Our data suggest that *0401 Tg mice generate a strong immune response to lipopolysaccharide and may be efficient in clearing infection, and may *0401 have been evolutionarily selected for this ability. Autoimmunity, such as RA, could likely be a bystander effect of the cytokine storm that, along with the presence of low Treg-cell numbers in *0401 Tg mice, causes immune dysregulation.


Subject(s)
Arthritis, Experimental/immunology , HLA-DRB1 Chains/immunology , T-Lymphocytes, Regulatory/cytology , Animals , Arthritis, Rheumatoid/immunology , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Female , HLA-DRB1 Chains/genetics , Inflammation/immunology , Interleukin-17/biosynthesis , Lipopolysaccharides/immunology , Male , Mice , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology
18.
Clin Immunol ; 152(1-2): 25-35, 2014.
Article in English | MEDLINE | ID: mdl-24631425

ABSTRACT

Individuals carrying DRB1*0401 who smoke cigarettes are at an increased risk of developing severe seropositive RA. To determine how cigarette smoke (CS) interacts with host genetic factors in the induction of RA-associated autoimmunity, we used transgenic mice carrying the RA-susceptible HLA genes DR4 and DQ8, but lacking all endogenous murine class II molecules. Cigarette smoke exposure augmented peptidylarginine deiminase (PAD) enzyme expression, and enhanced immune responses to citrullinated collagen and vimentin. Here we show for the first time that DQ molecules can present citrullinated peptides much more efficiently than native peptides. Interestingly, CS exposure suppressed collagen-induced arthritis (CIA) in DRB1*0401 mice although innate immune response was enhanced. On the other hand, CS exposure exacerbated CIA in DQ8 mice, which was accompanied by an increased expression of Th17 gene transcripts in lungs. These observations suggest that cigarette smoke promotes antigen-specific autoimmunity that is profoundly influenced by host genetic factors.


Subject(s)
Arthritis, Rheumatoid/immunology , HLA-DQ Antigens/genetics , HLA-DR4 Antigen/genetics , Smoke/adverse effects , Smoking/adverse effects , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/genetics , Autoimmunity/drug effects , Cell Proliferation/drug effects , Collagen/immunology , Cytokines/blood , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Genetic Predisposition to Disease , HLA-DQ Antigens/immunology , HLA-DR4 Antigen/immunology , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/immunology , Hydrolases/metabolism , Lung/cytology , Lung/immunology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Transgenic , Protein-Arginine Deiminases , Th17 Cells/drug effects , Th17 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Vimentin/immunology
19.
Article in English | MEDLINE | ID: mdl-26473171

ABSTRACT

Multiple Sclerosis (MS) is a chronic and debilitating disease of the central nervous system linked to both genetic and environmental factors. Among the genetic factors, MHC, especially HLA class-II, is strongly associated with predisposition to MS. Although in vitro studies have helped us understand some aspects of HLA class-II association with the disease, performing in vivo analysis is necessary in order to further understand this correlation. Studying the role of class-II genes in vivo is a difficult task due to the heterogeneity of human population, the complexity of MHC, and the strong linkage disequilibrium among different class-II genes. To overcome this challenge, we generated HLA class-II transgenic mice to study the role of these molecules in MS. Among the environmental factors linked with MS, ultra violet radiation (UVR)/vitamin-D is suggested to have protective effect against the development of the disease. Indeed, genetic studies have shown that presence of susceptible HLA-Class II and decrease in UVR exposure or vitamin D levels together increase risk of MS. Therefore, this study was designed to investigate the direct effect of UVR on immune response using novel humanized HLA-class II transgenic mice. HLA-class II transgenic mice expressing MS susceptible HLA-DR2 allele were treated with different doses of UVR (0.50-3.75 kJ/day) for seven consecutive days. T-cell proliferation, immune cell sub-populations and cytokines levels were analyzed. Our results show that treatment with UVR increased levels of regulatory CD4+FoxP3+ T cells and Gr1+ CD11b+ suppressive macrophages. Thus our study indicates that UVR modulates the immune response towards a tolerogenic phenotype in HLA-transgenic mice immunized with MOG35-55. Therefore, HLA class-II transgenic mice offer a novel tool to decipher the mechanism by which interaction between environmental and genetic factors play a role in predisposition and/or protection against development of MS.

20.
PLoS One ; 8(11): e78687, 2013.
Article in English | MEDLINE | ID: mdl-24236037

ABSTRACT

Human and animal studies strongly suggest that dietary gluten could play a causal role in the etiopathogenesis of type 1 diabetes (T1D). However, the mechanisms have not been elucidated. Recent reports indicate that the intestinal microbiome has a major influence on the incidence of T1D. Since diet is known to shape the composition of the intestinal microbiome, we investigated using non-obese diabetic (NOD) mice whether changes in the intestinal microbiome could be attributed to the pro- and anti-diabetogenic effects of gluten-containing and gluten-free diets, respectively. NOD mice were raised on gluten-containing chows (GCC) or gluten-free chows (GFC). The incidence of diabetes was determined by monitoring blood glucose levels biweekly using a glucometer. Intestinal microbiome composition was analyzed by sequencing 16S rRNA amplicons derived from fecal samples. First of all, GCC-fed NOD mice had the expected high incidence of hyperglycemia whereas NOD mice fed with a GFC had significantly reduced incidence of hyperglycemia. Secondly, when the fecal microbiomes were compared, Bifidobacterium, Tannerella, and Barnesiella species were increased (p = 0.03, 0.02, and 0.02, respectively) in the microbiome of GCC mice, where as Akkermansia species was increased (p = 0.02) in the intestinal microbiomes of NOD mice fed GFC. Thirdly, both of the gluten-free chows that were evaluated, either egg white based (EW-GFC) or casein based (C-GFC), significantly reduced the incidence of hyperglycemia. Interestingly, the gut microbiome from EW-GFC mice was similar to C-GFC mice. Finally, adding back gluten to the gluten-free diet reversed its anti-diabetogenic effect, reduced Akkermansia species and increased Bifidobacterium, Tannerella, and Barnesiella suggesting that the presence of gluten is directly responsible for the pro-diabetogenic effects of diets and it determines the gut microflora. Our novel study thus suggests that dietary gluten could modulate the incidence of T1D by changing the gut microbiome.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Diet, Gluten-Free , Glutens/adverse effects , Intestines/microbiology , Microbiota , Animals , Autoantibodies/blood , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/immunology , Dietary Proteins/adverse effects , Feces/microbiology , Female , Humans , Immunoglobulin G/blood , Incidence , Insulin/immunology , Male , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...