Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Immunity ; 57(7): 1648-1664.e9, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38876098

RESUMO

Allogeneic T cell expansion is the primary determinant of graft-versus-host disease (GVHD), and current dogma dictates that this is driven by histocompatibility antigen disparities between donor and recipient. This paradigm represents a closed genetic system within which donor T cells interact with peptide-major histocompatibility complexes (MHCs), though clonal interrogation remains challenging due to the sparseness of the T cell repertoire. We developed a Bayesian model using donor and recipient T cell receptor (TCR) frequencies in murine stem cell transplant systems to define limited common expansion of T cell clones across genetically identical donor-recipient pairs. A subset of donor CD4+ T cell clonotypes differentially expanded in identical recipients and were microbiota dependent. Microbiota-specific T cells augmented GVHD lethality and could target microbial antigens presented by gastrointestinal epithelium during an alloreactive response. The microbiota serves as a source of cognate antigens that contribute to clonotypic T cell expansion and the induction of GVHD independent of donor-recipient genetics.


Assuntos
Doença Enxerto-Hospedeiro , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/microbiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T CD4-Positivos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Microbiota/imunologia , Seleção Clonal Mediada por Antígeno , Transplante Homólogo , Teorema de Bayes , Transplante de Células-Tronco/efeitos adversos , Camundongos Endogâmicos BALB C , Microbioma Gastrointestinal/imunologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos
2.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33753483

RESUMO

Genome-wide association studies have identified ICOSLG, which encodes the inducible costimulator ligand (ICOSLG or ICOSL) as a susceptibility locus for inflammatory bowel disease. ICOSL has been implicated in the enhancement of pattern recognition receptor signaling in dendritic cells, induction of IL-10 production by CD4 T cells, and the generation of high-affinity antibodies to specific antigens-all of which can potentially explain its involvement in gastrointestinal inflammation. Here, we show that murine ICOSL deficiency results in significant enrichment of IL-10-producing CD4 T cells particularly in the proximal large intestine. Transient depletion of IL-10-producing cells from adult ICOSL-deficient mice induced severe colonic inflammation that was prevented when mice were first treated with metronidazole. ICOSL-deficient mice displayed reduced IgA and IgG antibodies in the colon mucus and impaired serum antibody recognition of microbial antigens, including flagellins derived from mucus-associated bacteria of the Lachnospiraceae family. Confirming the synergy between ICOSL and IL-10, ICOSL deficiency coupled with CD4-specific deletion of the Il10 gene resulted in juvenile onset colitis that was impeded when pups were fostered by ICOSL-sufficient dams. In this setting, we found that both maternally acquired and host-derived antibodies contribute to the life anti-commensal antibody repertoire that mediates this protection in early life. Collectively, our findings reveal a partnership between ICOSL-dependent anti-commensal antibodies and IL-10 in adaptive immune regulation of the microbiota in the large intestine. Furthermore, we identify ICOSL deficiency as an effective platform for exploring the functions of anti-commensal antibodies in host-microbiota mutualism.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Microbioma Gastrointestinal/imunologia , Ligante Coestimulador de Linfócitos T Induzíveis/metabolismo , Doenças Inflamatórias Intestinais/imunologia , Interleucina-10/metabolismo , Animais , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/imunologia , Linfócitos T CD4-Positivos/metabolismo , Colo/imunologia , Colo/microbiologia , Colo/patologia , Modelos Animais de Doenças , Feminino , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Ligante Coestimulador de Linfócitos T Induzíveis/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Interleucina-10/genética , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Knockout , Transdução de Sinais/imunologia , Simbiose/imunologia
3.
Gastroenterology ; 161(2): 522-535.e6, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33844987

RESUMO

BACKGROUND AND AIMS: Crohn's disease and ulcerative colitis are characterized by dysregulated adaptive immune responses to the microbiota in genetically susceptible individuals, but the specificity of these responses remains largely undefined. Therefore, we developed a microbiota antigen microarray to characterize microbial antibody reactivity, particularly to human-derived microbiota flagellins, in inflammatory bowel disease. METHODS: Sera from healthy volunteers (n = 87) at the University of Alabama at Birmingham and from patients recruited from the Kirklin Clinic of University of Alabama at Birmingham Hospital, including patients with Crohn's disease (n = 152) and ulcerative colitis (n = 170), were individually probed against microbiota bacterial flagellins of both mouse and human origin and analyzed for IgG and IgA antibody responses. Circulating flagellin-reactive T effector (CD4+CD154+) and T regulatory (CD4+CD137+) cells were isolated and evaluated in selected patients. Resulting adaptive immune responses were compared with corresponding clinical data to determine relevancy to disease behavior. RESULTS: We show that patients with IBD express selective patterns of antibody reactivity to microbiota flagellins. Patients with Crohn's disease, but not patients with ulcerative colitis, display augmented serum IgG to human ileal-localized Lachnospiraceae flagellins, with a subset of patients having high responses to more than 10 flagellins. Elevated responses to CBir1, a mouse Lachnospiraceae flagellin used clinically to diagnose CD, correlated with multi-Lachnospiraceae flagellin reactivity. In this subset of patients with CD, multi-flagellin reactivity was associated with elevated flagellin-specific CD154+CD45RA- T memory cells, a reduced ratio of flagellin-reactive CD4+ T regulatory to T effector cells, and a high frequency of disease complications. CONCLUSIONS: Patients with Crohn's disease display strong adaptive immune response to human-derived Lachnospiraceae flagellins, which may be targeted for prognosis and future personalized therapies.


Assuntos
Imunidade Adaptativa , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Linfócitos T CD4-Positivos/imunologia , Clostridiales/imunologia , Doença de Crohn/imunologia , Flagelina/imunologia , Imunoglobulina G/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos de Bactérias/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/microbiologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/microbiologia , Estudos de Casos e Controles , Clostridiales/metabolismo , Colite Ulcerativa/sangue , Colite Ulcerativa/imunologia , Colite Ulcerativa/microbiologia , Doença de Crohn/sangue , Doença de Crohn/microbiologia , Estudos Transversais , Feminino , Flagelina/metabolismo , Humanos , Imunoglobulina A/sangue , Masculino , Pessoa de Meia-Idade , Adulto Jovem
4.
Clin Immunol ; 216: 108463, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32437923

RESUMO

OBJECTIVES: Patients with Crohn's disease often produce antibodies against flagellated intestinal bacteria. There are mixed data as to whether such antibodies are present in patients with spondyloarthritis. Our objectives were to evaluate for the presence of antibodies against intestinal organisms in children with enthesitis related arthritis (ERA). METHODS: Children with ERA and healthy controls were recruited at three sites. Sera were plated on a nitrocellulose array and incubated with labelled antibodies to human IgA and IgG. RESULTS: At UAB, patients and controls had similar antibody levels against the majority of the bacteria selected, with the exception of increased IgA antibodies among ERA patients against Prevotella oralis (1231 [IQR 750, 2566] versus 706 [IQR 428, 1106], p = .007.) These findings were partially validated at a second but not at a third site. CONCLUSIONS: ERA patients may produce increased IgA antibodies against P. oralis. The possible significance of this finding bears further exploration.


Assuntos
Antígenos de Bactérias/sangue , Antígenos de Bactérias/imunologia , Artrite Juvenil/sangue , Artrite Juvenil/imunologia , Prevotella/imunologia , Artrite Juvenil/microbiologia , Criança , Doença de Crohn/imunologia , Doença de Crohn/microbiologia , Feminino , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Masculino
5.
Immunity ; 34(3): 293-302, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21435584

RESUMO

Inflammatory bowel disease appears to result from an abnormal host immune response to the intestinal microbiota. Experimental models have allowed the dissection of the complex dialog between the host and its microbiota. Through genetic manipulation of the host genome the immune compartments, cells, molecules, and genes that are critical for maintenance of intestinal homeostasis are being identified. Genetic association studies in humans have identified over 100 susceptibility loci. Although there is remarkable coherence between the experimental model and the human genetic data, a full understanding of the mechanisms involved in genetic susceptibility to IBD and of gene-gene and gene-environmental interactions will require a "next generation" of experimental models.


Assuntos
Colite/imunologia , Interações Hospedeiro-Patógeno , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Metagenoma/fisiologia , Animais , Modelos Animais de Doenças , Homeostase , Humanos
6.
Genes Immun ; 20(2): 158-166, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29599513

RESUMO

Studies have identified abnormalities in the microbiota of patients with arthritis. To evaluate the pathogenicity of human microbiota, we performed fecal microbial transplantation from children with spondyloarthritis and controls to germ-free KRN/B6xNOD mice. Ankle swelling was equivalent in those that received patient vs. control microbiota. Principal coordinates analysis revealed incomplete uptake of the human microbiota with over-representation of two genera (Bacteroides and Akkermansia) among the transplanted mice. The microbiota predicted the extent of ankle swelling (R2 = 0.185, p = 0.018). The abundances of Bacteroides (r = -0.510, p = 0.010) inversely and Akkermansia (r = 0.367, p = 0.078) directly correlated with ankle swelling. Addition of Akkermansia muciniphila to Altered Schaedler's Flora (ASF) resulted in small but statistically significant increased ankle swelling as compared to mice that received ASF alone (4.0 mm, 3.9-4.1 vs. 3.9 mm, IQR 3.6-4.0, p = 0.041), as did addition of A. muciniphila cultures to transplanted human microbiota as compared to mice that received transplanted human microbiota alone (4.5 mm, IQR 4.3-5.5 vs. 4.1 mm, IQR 3.9-4.3, p = 0.019). This study supports previous findings of an association between A. muciniphila and arthritis.


Assuntos
Artrite/microbiologia , Microbioma Gastrointestinal , Adolescente , Animais , Tornozelo/patologia , Bacteroides/isolamento & purificação , Bacteroides/patogenicidade , Criança , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Verrucomicrobia/isolamento & purificação , Verrucomicrobia/patogenicidade
7.
PLoS Pathog ; 13(1): e1006087, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28125732

RESUMO

HIV-1 infection is associated with an early and profound depletion of mucosal memory CD4+ T cells, a population that plays an indispensable role in the regulation of isotype switching and transepithelial transport of antibodies. In this study, we addressed whether the depletion of CD4+ T cell in HIV-1-infected individuals results in altered humoral responses specific to antigens encountered at mucosal surfaces. Comprehensive protein microarray of systemic humoral responses to intestinal microbiota demonstrated reduced IgG responses to antigens derived from Proteobacteria and Firmicutes but not Bacteroidetes. Importantly, intestinal secretions of antiretroviral therapy-treated HIV-1-infected individuals exhibited a significant elevation of IgM levels and decreased IgA/IgM and IgG/IgM ratios of antibodies specific to a variety of microbial and food antigens. The presented findings indicate reduced competence of mucosal B cells for class switch recombination from IgM to other isotypes limiting their capacity to react to changing antigenic variety in the gut lumen. Decreased availability of microbiota-specific IgA and IgG may be an important factor contributing to the translocation of microbial antigens across the intestinal mucosal barrier and their systemic dissemination that drives chronic inflammation in HIV-1-infected individuals.


Assuntos
Antígenos/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Microbiota/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Doença Crônica , Alimentos , Regulação da Expressão Gênica , Infecções por HIV/virologia , Humanos , Imunidade Humoral , Imunoglobulina A/imunologia , Switching de Imunoglobulina , Imunoglobulina G/imunologia , Inflamação , Mucosa Intestinal/imunologia , Mucosa Intestinal/virologia
8.
J Immunol ; 199(1): 312-322, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28539431

RESUMO

Th17 cells play a role as an inflammation mediator in a variety of autoimmune disorders, including inflammatory bowel disease, and thus are widely considered to be pathogenic. However, Th17 cells are present in the normal intestine and show a homeostatic phenotype; that is, they participate in the maintenance of intestinal homeostasis rather than inducing inflammation. We observed an enlarged Th17 population in the small intestine of C57BL/6.IgA-/- mice compared with wild-type mice, which was further amplified with cholera toxin (CT) immunization without causing intestinal inflammation. The increased Th17 induction and the correspondingly 10-fold higher CT B subunit-specific serum IgG response in IgA-/- mice after CT immunization was microbiota dependent and was associated with increased segmented filamentous bacteria in the small intestine of IgA-/- mice. Oral administration of vancomycin greatly dampened both CT immunogenicity and adjuvanticity, and the differential CT responses in IgA-/- and wild-type mice disappeared after intestinal microbiota equalization. Using gnotobiotic mouse models, we found that CT induction of homeostatic intestinal Th17 responses was supported not only by segmented filamentous bacteria, but also by other commensal bacteria. Furthermore, transcriptome analysis using IL-17AhCD2 reporter mice revealed a similar gene expression profile in CT-induced intestinal Th17 cells and endogenous intestinal Th17 cells at homeostasis, with upregulated expression of a panel of immune-regulatory genes, which was distinctly different from the gene expression profile of pathogenic Th17 cells. Taken together, we identified a nonpathogenic signature of intestinal homeostatic Th17 cells, which are actively regulated by the commensal microbiota and can be selectively stimulated by CT.


Assuntos
Toxina da Cólera/imunologia , Microbioma Gastrointestinal/imunologia , Homeostase , Mucosa Intestinal/imunologia , Intestino Delgado/imunologia , Células Th17/imunologia , Animais , Toxina da Cólera/administração & dosagem , Microbioma Gastrointestinal/fisiologia , Perfilação da Expressão Gênica , Vida Livre de Germes , Deficiência de IgA/imunologia , Imunoglobulina A/imunologia , Doenças Inflamatórias Intestinais , Mucosa Intestinal/microbiologia , Intestino Delgado/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Vancomicina/administração & dosagem
9.
J Pediatr Gastroenterol Nutr ; 68(4): 502-508, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30540709

RESUMO

OBJECTIVES: The gut microbiome is believed to play a role in the susceptibility to and treatment of Clostridium difficile infections (CDIs). It is, however, unknown whether the gut microbiome is also affected by asymptomatic C difficile colonization. Our study aimed to evaluate the fecal microbiome of children based on C difficile colonization, and CDI risk factors, including antibiotic use and comorbid inflammatory bowel disease (IBD). METHODS: Subjects with IBD and non-IBD controls were prospectively enrolled from pediatric clinics for a biobanking project (n = 113). A fecal sample was collected from each subject for research purposes only and was evaluated for asymptomatic toxigenic C difficile colonization. Fecal microbiome composition was determined by 16S rRNA sequencing. RESULTS: We found reduced bacterial diversity and altered microbiome composition in subjects with C difficile colonization, concurrent antibiotic use, and/or concomitant IBD (all P < 0.05). Accounting for antibiotic use and IBD status, children colonized with C difficile had significant enrichment in taxa from the genera Ruminococcus, Eggerthella, and Clostridium. Children without C difficile had increased relative abundances of Faecalibacterium and Rikenellaceae. Imputed metagenomic functions of those colonized were enriched for genes in oxidative phosphorylation and beta-lactam resistance, whereas in the subjects without C difficile, several functions in translation and metabolism were over-represented. CONCLUSIONS: In children, C difficile colonization, or factors that predispose to colonization such as antibiotic use and IBD status were associated with decreased gut bacterial diversity and altered microbiome composition. Averting such microbiome alterations may be a method to prevent or treat CDI.


Assuntos
Clostridioides difficile , Infecções por Clostridium/microbiologia , Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais/complicações , Adolescente , Alabama , Baltimore , Criança , Pré-Escolar , Fezes/microbiologia , Feminino , Humanos , Masculino , Estudos Prospectivos , Adulto Jovem
10.
Nature ; 498(7452): 113-7, 2013 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-23698371

RESUMO

Innate lymphoid cells (ILCs) are a recently characterized family of immune cells that have critical roles in cytokine-mediated regulation of intestinal epithelial cell barrier integrity. Alterations in ILC responses are associated with multiple chronic human diseases, including inflammatory bowel disease, implicating a role for ILCs in disease pathogenesis. Owing to an inability to target ILCs selectively, experimental studies assessing ILC function have predominantly used mice lacking adaptive immune cells. However, in lymphocyte-sufficient hosts ILCs are vastly outnumbered by CD4(+) T cells, which express similar profiles of effector cytokines. Therefore, the function of ILCs in the presence of adaptive immunity and their potential to influence adaptive immune cell responses remain unknown. To test this, we used genetic or antibody-mediated depletion strategies to target murine ILCs in the presence of an adaptive immune system. We show that loss of retinoic-acid-receptor-related orphan receptor-γt-positive (RORγt(+)) ILCs was associated with dysregulated adaptive immune cell responses against commensal bacteria and low-grade systemic inflammation. Remarkably, ILC-mediated regulation of adaptive immune cells occurred independently of interleukin (IL)-17A, IL-22 or IL-23. Genome-wide transcriptional profiling and functional analyses revealed that RORγt(+) ILCs express major histocompatibility complex class II (MHCII) and can process and present antigen. However, rather than inducing T-cell proliferation, ILCs acted to limit commensal bacteria-specific CD4(+) T-cell responses. Consistent with this, selective deletion of MHCII in murine RORγt(+) ILCs resulted in dysregulated commensal bacteria-dependent CD4(+) T-cell responses that promoted spontaneous intestinal inflammation. These data identify that ILCs maintain intestinal homeostasis through MHCII-dependent interactions with CD4(+) T cells that limit pathological adaptive immune cell responses to commensal bacteria.


Assuntos
Bactérias/imunologia , Linfócitos T CD4-Positivos/imunologia , Imunidade Inata/imunologia , Intestinos/imunologia , Intestinos/microbiologia , Simbiose , Animais , Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/patologia , Proliferação de Células , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Inflamação/patologia , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Interleucinas/metabolismo , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Interleucina 22
12.
Immunology ; 154(1): 28-37, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29338074

RESUMO

Host-microbiota mutualism has been established during long-term co-evolution. A diverse and rich gut microbiota plays an essential role in the development and maturation of the host immune system. Education of the adaptive immune compartment by gut microbiota antigens is important in establishing immune balance. In particular, a critical time frame immediately after birth provides a 'window of opportunity' for the development of lymphoid structures, differentiation and maturation of T and B cells and, most importantly, establishment of immune tolerance to gut commensals. Depending on the colonization niche, antigen type and metabolic property of different gut microbes, CD4 T-cell responses vary greatly, which results in differentiation into distinct subsets. As a consequence, certain bacteria elicit effector-like immune responses by promoting the production of pro-inflammatory cytokines such as interferon-γ and interleukin-17A, whereas other bacteria favour the generation of regulatory CD4 T cells and provide help with gut homeostasis. The microbiota have profound effects on B cells also. Gut microbial exposure leads to a continuous diversification of B-cell repertoire and the production of T-dependent and -independent antibodies, especially IgA. These combined effects of the gut microbes provide an elegant educational process to the adaptive immune network. Contrariwise, failure of this process results in a reduced homeostasis with the gut microbiota, and an increased susceptibility to various immune disorders, both inside and outside the gut. With more definitive microbial-immune relations waiting to be discovered, modulation of the host gut microbiota has a promising future for disease intervention.


Assuntos
Imunidade Adaptativa , Antígenos de Bactérias/imunologia , Bactérias/imunologia , Microbioma Gastrointestinal , Intestinos/imunologia , Intestinos/microbiologia , Animais , Antígenos de Bactérias/metabolismo , Linfócitos B/imunologia , Linfócitos B/microbiologia , Bactérias/metabolismo , Disbiose , Interações Hospedeiro-Patógeno , Humanos , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/microbiologia , Mucosa Intestinal/metabolismo , Linfócitos T/imunologia , Linfócitos T/microbiologia
13.
Immunity ; 30(1): 92-107, 2009 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-19119024

RESUMO

Development of T helper (Th) 17 cells requires transforming growth factor (TGF)-beta and interleukin (IL)-6 and is independent of the Th1 pathway. Although T cells that produce interferon (IFN)-gamma are a recognized feature of Th17 cell responses, mice deficient for STAT4 and T-bet-two prototypical Th1 transcription factors-are protected from autoimmunity associated with Th17 pathogenesis. To examine the fate and pathogenic potential of Th17 cells and origin of IFN-gamma-producing T cells that emerge during Th17 immunity, we developed IL-17F reporter mice that identify cells committed to expression of IL-17F and IL-17A. Th17 cells required TGF-beta for sustained expression of IL-17F and IL-17A. In the absence of TGF-beta, both IL-23 and IL-12 acted to suppress IL-17 and enhance IFN-gamma production in a STAT4- and T-bet-dependent manner, albeit with distinct efficiencies. These results support a model of late Th17 developmental plasticity with implications for autoimmunity and host defense.


Assuntos
Linhagem da Célula/imunologia , Interleucina-17/metabolismo , Subpopulações de Linfócitos T/imunologia , Células Th1/imunologia , Células Th2/imunologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Células Cultivadas , Citometria de Fluxo , Interferon gama/metabolismo , Interleucina-23/metabolismo , Camundongos , Camundongos Knockout
14.
Nature ; 489(7415): 231-41, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-22972296

RESUMO

The emergence of the adaptive immune system in vertebrates set the stage for evolution of an advanced symbiotic relationship with the intestinal microbiota. The defining features of specificity and memory that characterize adaptive immunity have afforded vertebrates the mechanisms for efficiently tailoring immune responses to diverse types of microbes, whether to promote mutualism or host defence. These same attributes can put the host at risk of immune-mediated diseases that are increasingly linked to the intestinal microbiota. Understanding how the adaptive immune system copes with the remarkable number and diversity of microbes that colonize the digestive tract, and how the system integrates with more primitive innate immune mechanisms to maintain immune homeostasis, holds considerable promise for new approaches to modulate immune networks to treat and prevent disease.


Assuntos
Intestinos/imunologia , Intestinos/microbiologia , Metagenoma/imunologia , Antígenos de Grupos Sanguíneos/imunologia , Cesárea , Epitélio/imunologia , Feminino , Homeostase/imunologia , Humanos , Lactente , Recém-Nascido , Gravidez , Linfócitos T/imunologia , Vagina/microbiologia
15.
Immunol Rev ; 260(1): 206-20, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24942691

RESUMO

The human host has coevolved with the collective of bacteria species, termed microbiota, in a complex fashion that affects both innate and adaptive immunity. Differential regulation of regulatory T-cell and effector T-cell responses are a direct result of specific microbial species present within the gut, and this relationship is subject to dysregulation during inflammation and disease. The microbiota varies widely between individuals and has a profound effect on how one reacts to various environmental stimuli, particularly if a person is genetically predisposed to an immune-mediated inflammatory disorder such as inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC). Approximately, half of all CD patients have elevated antibodies to CBir1, a microbiota flagellin common to mice and humans, demonstrating flagellins as immunodominant antigens in the intestines. This review focuses on the use of flagellins as probes to study microbiota-specific responses in the context of health and disease as well as probes of innate and adaptive responses employed by the host to deal with the overwhelming bacterial presence of the microbiota.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Microbiota/imunologia , Animais , Flagelina/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/microbiologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
16.
Eur J Immunol ; 46(5): 1162-7, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26840377

RESUMO

It has been shown that while commensal bacteria promote Th1, Th17 and Treg cells in lamina propria (LP) in steady-state conditions, they suppress mucosal Th2 cells. However, it is still unclear whether there are specific commensal organisms down-regulating Th2 responses, and the mechanism involved. Here we demonstrate that commensal A4 bacteria, a member of the Lachnospiraceae family, which produce an immunodominant microbiota CBir1 antigen, inhibits LP Th2-cell development. When transferred into the intestines of RAG(-/-) mice, CBir1-specific T cells developed predominately towards Th1 cells and Th17 cells, but to a lesser extent into Th2 cells. The addition of A4 bacterial lysates to CD4(+) T-cell cultures inhibited production of IL-4. A4 bacteria stimulated dendritic cell production of TGF-ß, and blockade of TGF-ß abrogated A4 bacteria inhibition of Th2-cell development in vitro and in vivo. Collectively, our data show that A4 bacteria inhibit Th2-cell differentiation by inducing dendritic cell production of TGF-ß.


Assuntos
Células Dendríticas/imunologia , Bactérias Gram-Positivas/imunologia , Mucosa/imunologia , Simbiose , Células Th2/imunologia , Fator de Crescimento Transformador beta/imunologia , Animais , Diferenciação Celular , Células Cultivadas , Bactérias Gram-Positivas/química , Interleucina-4/biossíntese , Interleucina-4/imunologia , Ativação Linfocitária , Camundongos , Mucosa/microbiologia , Células Th1/imunologia , Células Th17/imunologia , Células Th2/fisiologia , Fator de Crescimento Transformador beta/biossíntese
17.
Eur J Immunol ; 45(4): 1010-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25605286

RESUMO

Differentiated CD4(+) T cells preserve plasticity under various conditions. However, the stability of Th1 cells is unclear, as is whether Th1 cells can convert into Th17 cells and thereby contribute to the generation of IFN-γ(+) IL-17(+) CD4(+) T cells, the number of which correlates with severity of colitis. We investigated whether IFN-γ(+) Th1 cells can convert into Th17 cells under intestinal inflammation and the mechanisms involved. IFN-γ(Thy1.1+) Th1 cells were generated by culturing naïve CD4(+) T cells from IFN-γ(Thy1.1) CBir1 TCR-Tg reporter mice, whose TCR is specific for an immunodominant microbiota antigen, CBir1 flagellin, under Th1 polarizing conditions. IFN-γ(Thy1.1+) Th1 cells induced colitis in Rag(-/-) mice after adoptive transfer and converted into IL-17(+) Th17, but not Foxp3(+) Treg cells in the inflamed intestines. TGF-ß and IL-6, but not IL-1ß and IL-23, regulated Th1 conversion into Th17 cells. TGF-ß induction of transcriptional factor Runx1 is crucial for the conversion, since silencing Runx1 by siRNA inhibited Th1 conversion into Th17 cells. Furthermore, TGF-ß enhanced histone H3K9 acetylation but inhibited H3K9 trimethylation of Runx1- and ROR-γt-binding sites on il-17 or rorc gene in Th1 cells. We conclude that Th1 cells convert into Th17 cells under inflammatory conditions in intestines, which is possibly mediated by TGF-ß induction of Runx1.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/biossíntese , Mucosa Intestinal/imunologia , Células Th1/citologia , Células Th17/citologia , Fator de Crescimento Transformador beta/metabolismo , Acetilação , Animais , Sítios de Ligação , Diferenciação Celular/imunologia , Células Cultivadas , Colite/imunologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Flagelina/imunologia , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Interferon gama/biossíntese , Interleucina-17/biossíntese , Interleucina-1beta/metabolismo , Interleucina-2/metabolismo , Interleucina-23/metabolismo , Interleucina-6/metabolismo , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/biossíntese , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/biossíntese , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Células Th1/imunologia , Células Th17/imunologia
18.
J Allergy Clin Immunol ; 136(5): 1378-86.e1-5, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26014812

RESUMO

BACKGROUND: Although immune responses directed against antigens from the intestinal microbiota are observed in certain diseases, the normal human adaptive immune response to intestinal microbiota is poorly defined. OBJECTIVE: Our goal was to assess the adaptive immune response to the intestinal microbiota present in 143 healthy adults and compare this response with the response observed in 52 children and their mothers at risk of having allergic disease. METHODS: Human serum was collected from adults and children followed from birth to 7 years of age, and the serum IgG response to a panel of intestinal microbiota antigens was assessed by using a novel protein microarray. RESULTS: Nearly every subject tested, regardless of health status, had serum IgG that recognized a common set of antigens. Seroreactivity to the panel of antigens was significantly lower in atopic adults. Healthy infants expressed the highest level of IgG seroreactivity to intestinal microbiota antigens. This adaptive response developed between 6 and 12 months of age and peaked around 2 years of age. Low IgG responses to certain clusters of microbiota antigens during infancy were associated with allergy development during childhood. CONCLUSIONS: There is an observed perturbation of the adaptive response to antigens from the microbiota in allergic subjects. These perturbations are observable even in childhood, suggesting that optimal stimulation of the adaptive immune system by the microbiota might be needed to prevent certain immune-mediated diseases.


Assuntos
Antígenos de Bactérias/imunologia , Microbioma Gastrointestinal/imunologia , Hipersensibilidade/imunologia , Intestinos/imunologia , Imunidade Adaptativa , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Imunoglobulina G/sangue , Lactente , Recém-Nascido , Intestinos/microbiologia , Masculino , Análise em Microsséries
19.
Eur J Immunol ; 44(3): 673-82, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24293139

RESUMO

Commensal flora plays an important role in the development of the mucosal immune system and in maintaining intestinal homeostasis. However, the mechanisms involved in regulation of host-microbiota interaction are still not completely understood. In this study, we examined how microbiota and intestinal inflammatory conditions regulate host microRNA expression and observed lower microRNA-107 (miR-107) expression in the inflamed intestines of colitic mice, compared with that in normal control mice. miR-107 was predominantly reduced in epithelial cells and CD11c(+) myeloid cells including dendritic cells and macrophages in the inflamed intestines. We demonstrate that IL-6, IFN-γ, and TNF-α downregulated, whereas TGF-ß promoted, miR-107 expression. In addition, miR-107 expression was higher in the intestines of germ-free mice than in mice housed under specific pathogen-free conditions, and the presence of microbiota downregulated miR-107 expression in DCs and macrophages in a MyD88- and NF-κB-dependent manner. We determined that the ectopic expression of miR-107 specifically repressed the expression of IL-23p19, a key molecule in innate immune responses to commensal bacteria. We concluded that regulation of miR-107 by intestinal microbiota and proinflammatory cytokine serve as an important pathway for maintaining intestinal homeostasis.


Assuntos
Subunidade p19 da Interleucina-23/genética , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , MicroRNAs/genética , Microbiota , Células Mieloides/metabolismo , Animais , Bactérias/imunologia , Bactérias/metabolismo , Pareamento de Bases , Sequência de Bases , Colite/genética , Colite/imunologia , Citocinas/metabolismo , Citocinas/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/farmacologia , Subunidade p19 da Interleucina-23/química , Subunidade p19 da Interleucina-23/metabolismo , Intestinos/imunologia , Ligantes , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , MicroRNAs/química , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Receptores Toll-Like/metabolismo
20.
Dig Dis ; 33(2): 131-136, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25925913

RESUMO

The comprehensive collection of bacterial species, termed microbiota, within human and other mammalian hosts has profound effects on both innate and adaptive immunity. Multiple host innate mechanisms contribute to intestinal homeostasis, including epithelial production of protective mucin layers maintaining spatial segregation in the intestine as well as epithelial cell secretion of a broad range of antimicrobial peptides. Additionally, epithelial cells employ autophagy to contain and eliminate invading bacteria; interestingly, genetic variants in specific autophagy genes are linked to susceptibility to Crohn's disease. Innate lymphoid cells, which rapidly respond to cytokine and microbial signals, have emerged as important regulators of the intestinal immune response to the microbiota. With regard to adaptive immunity, specific microbial species stimulate induction of regulatory T cells while others induce effector T cells within the gut. Such stimulation is subject to dysregulation during inflammation and disease, contributing to 'dysbiosis' or an abnormal microbiota composition that has been associated with a variety of immune-mediated inflammatory disorders, including celiac disease. The microbiota communicates with the immune system and vice versa; thus, an abnormal microbiota composition likely translates into an altered host immune response, though the exact mechanisms of such are not yet clear. Immunoglobulin A plays a critical role in limiting bacterial access to the host and in maintaining mutualism with the microbiota. Perturbation of the mucosal barrier via infection or other means can induce effector T cells reactive to the intestinal microbiota, and these cells can persist as memory cells for extended periods of time and potentially serve as pathogenic effector cells upon re-encounter with antigen. Health is associated with a diverse microbiota that functions to maintain the balance between T effector and T regulatory cells in the intestine. Whether dysbiosis can be reversed in immune-mediated disease, thus restoring health, is a question of intense interest for this active area of research.


Assuntos
Microbioma Gastrointestinal , Intestinos/microbiologia , Imunidade Adaptativa , Animais , Microbioma Gastrointestinal/imunologia , Humanos , Imunidade Inata , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestinos/imunologia , Linfócitos/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa