Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
2.
Infect Immun ; 90(2): e0022221, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34978927

RESUMO

Hypoxia-inducible transcription factor 1 (HIF-1) has been shown to enhance microbial killing and ameliorate the course of bacterial infections. While the impact of HIF-1 on inflammatory diseases of the gut has been studied intensively, its function in bacterial infections of the gastrointestinal tract remains largely elusive. With the help of a publicly available gene expression data set, we inferred significant activation of HIF-1 after oral infection of mice with Salmonella enterica serovar Typhimurium. Immunohistochemistry and Western blot analyses confirmed marked HIF-1α protein stabilization, especially in the intestinal epithelium. This prompted us to analyze conditional Hif1a-deficient mice to examine cell type-specific functions of HIF-1 in this model. Our results demonstrate enhanced noncanonical induction of HIF-1 activity upon Salmonella infection in the intestinal epithelium as well as in macrophages. Surprisingly, Hif1a deletion in intestinal epithelial cells did not impact inflammatory gene expression, bacterial spread, or disease outcomes. In contrast, Hif1a deletion in myeloid cells enhanced intestinal Cxcl2 expression and reduced the cecal Salmonella load. In vitro, HIF-1α-deficient macrophages showed overall impaired transcription of mRNA encoding proinflammatory factors; however, the intracellular survival of Salmonella was not impacted by HIF-1α deficiency.


Assuntos
Infecções por Salmonella , Salmonella typhimurium , Animais , Células Epiteliais/microbiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Mucosa Intestinal/microbiologia , Macrófagos , Camundongos , Infecções por Salmonella/genética , Salmonella typhimurium/genética
3.
J Immunol ; 202(1): 260-267, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30487173

RESUMO

Salmonella infection is a globally important cause of gastroenteritis and systemic disease and is a useful tool to study immune responses in the intestine. Although mechanisms leading to immune responses against Salmonella have been extensively studied, questions remain about how bacteria travel from the intestinal mucosa to the mesenteric lymph nodes (MLN), a key site for Ag presentation. In this study, we used a mouse model of infection with Salmonella enterica serovar Typhimurium (STM) to identify changes in intestinal immune cells induced during early infection. We then used fluorescently labeled STM to identify interactions with immune cells from the site of infection through migration in lymph to the MLN. We show that viable STM can be carried in the lymph by any subset of migrating dendritic cells but not by macrophages. Moreover, approximately half of the STM in lymph are not associated with cells at all and travel autonomously. Within the MLN, STM associates with dendritic cells and B cells but predominantly with MLN-resident macrophages. In conclusion, we describe the routes used by STM to spread systemically in the period immediately postinfection. This deeper understanding of the infection process could open new avenues for controlling it.


Assuntos
Células Dendríticas/imunologia , Mucosa Intestinal/microbiologia , Linfonodos/microbiologia , Macrófagos/imunologia , Mesentério/imunologia , Salmonella typhi/fisiologia , Febre Tifoide/imunologia , Animais , Células Dendríticas/microbiologia , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Humanos , Mucosa Intestinal/imunologia , Linfonodos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Febre Tifoide/microbiologia
4.
Immunology ; 159(1): 1-3, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31777065

RESUMO

The field of mucosal immunology has, for the last 10 years, been largely dominated by advances in our understanding of the commensal microbiota. Developments of novel experimental methodologies and analysis techniques have provided unparalleled insight into the profound impact the microbiota has on the development and function of the immune system. In this cross-journal review series published in Immunology and Clinical and Experimental Immunology, we aim to summarize the current state of research concerning the interplay between the microbiota and mucosal immunity. In addition, the series examines how the increased understanding of the microbiota is changing the nature of immunological research, both in the laboratory and in the clinic.


Assuntos
Microbioma Gastrointestinal/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Imunidade nas Mucosas , Animais , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia
5.
Trends Immunol ; 37(5): 287-296, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27066758

RESUMO

Starting at birth, the intestinal microbiota changes dramatically from a highly individual collection of microorganisms, dominated by comparably few species, to a mature, competitive, and diverse microbial community. Microbial colonization triggers and accompanies the maturation of the mucosal immune system and ultimately results in a mutually beneficial host-microbe interrelation in the healthy host. Here, we discuss the role of secretory immunoglobulin A (SIgA) during the establishment of the infant microbiota and life-long host-microbial homeostasis. We critically review the published literature on how SIgA affects the enteric microbiota and highlight the accessibility of the infant microbiota to therapeutic intervention.


Assuntos
Microbioma Gastrointestinal , Homeostase , Imunidade nas Mucosas , Imunoglobulina A/imunologia , Microbiota , Animais , Terapia Biológica , Humanos , Lactente , Recém-Nascido
6.
Trends Immunol ; 35(6): 270-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24794393

RESUMO

Mononuclear phagocytes (MPs) in the murine intestine, comprising dendritic cells (DCs) and macrophages (Mϕs), perform disparate yet complementary immunological functions. Functional analyses of these distinct MP subsets have been complicated by the substantial overlap in their surface phenotypes. Here, we review recent findings that have enabled more accurate definition of these MP subsets. We discuss these recent advances in the context of the current understanding of the functions of DCs and Mϕs in the maintenance of intestinal homeostasis, and how their functions may alter when homeostasis is disrupted.


Assuntos
Células Dendríticas/imunologia , Intestinos/imunologia , Macrófagos/imunologia , Animais , Diferenciação Celular , Células Dendríticas/metabolismo , Homeostase , Humanos , Imunidade , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Macrófagos/metabolismo , Fagócitos/imunologia , Fagócitos/metabolismo , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
8.
J Immunol ; 192(5): 2442-8, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24470502

RESUMO

CD4(+) T cells have long been grouped into distinct helper subsets on the basis of their cytokine-secretion profile. In recent years, several subsets of innate lymphoid cell have been described as key producers of these same Th-associated cytokines. However, the functional relationship between Th cells and innate lymphoid cells (ILCs) remains unclear. We show in this study that lineage-negative ST2(+)ICOS(+)CD45(+) type 2 ILCs and CD4(+) T cells can potently stimulate each other's function via distinct mechanisms. CD4(+) T cell provision of IL-2 stimulates type 2 cytokine production by type 2 ILCs. By contrast, type 2 ILCs modulate naive T cell activation in a cell contact-dependent manner, favoring Th2 while suppressing Th1 differentiation. Furthermore, a proportion of type 2 ILCs express MHC class II and can present peptide Ag in vitro. Importantly, cotransfer experiments show that type 2 ILCs also can boost CD4(+) T cell responses to Ag in vivo.


Assuntos
Antígenos de Diferenciação/imunologia , Diferenciação Celular/imunologia , Citocinas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Imunidade Inata/fisiologia , Células Th2/imunologia , Animais , Antígenos de Diferenciação/genética , Diferenciação Celular/genética , Citocinas/genética , Antígenos de Histocompatibilidade Classe II/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Th1/citologia , Células Th1/imunologia , Células Th2/citologia
9.
J Immunol ; 193(1): 400-11, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24890717

RESUMO

Chemokine-directed leukocyte migration is crucial for effective immune and inflammatory responses. Conventional chemokine receptors (cCKRs) directly control cell movement; atypical chemokine receptors (ACKRs) regulate coexpressed cCKRs; and both cCKRs and ACKRs internalize chemokines to limit their abundance in vivo, a process referred to as scavenging. A leukocyte's migratory and chemokine-scavenging potential is determined by which cCKRs and ACKRs it expresses, and by the ligand specificity, signaling properties, and chemokine internalization capacity of these receptors. Most chemokines can bind at least one cCKR and one ACKR. CCL2 can bind to CCR2 (a cCKR) and two ACKRs (ACKR1 and ACKR2). In this study, by using fluorescent CCL2 uptake to label cells bearing functional CCL2 receptors, we have defined the expression profile, scavenging activity, and ligand specificity of CCL2 receptors on mouse leukocytes. We show that qualitative and quantitative differences in the expression of CCR2 and ACKR2 endow individual leukocyte subsets with distinctive CCL2 receptor profiles and CCL2-scavenging capacities. We reveal that some cells, including plasmacytoid dendritic cells, can express both CCR2 and ACKR2; that Ly6C(high) monocytes have particularly strong CCL2-scavenging potential in vitro and in vivo; and that CCR2 is a much more effective CCL2 scavenger than ACKR2. We confirm the unique, overlapping, ligand specificities of CCR2 and ACKR2 and, unexpectedly, find that cell context influences the interaction of CCL7 and CCL12 with CCR2. Fluorescent chemokine uptake assays were instrumental in providing these novel insights into CCL2 receptor biology, and the sensitivity, specificity, and versatility of these assays are discussed.


Assuntos
Quimiocina CCL2/imunologia , Células Dendríticas/imunologia , Monócitos/imunologia , Plasmócitos/imunologia , Receptores de Quimiocinas/imunologia , Animais , Quimiocina CCL2/genética , Quimiocina CCL7/genética , Quimiocina CCL7/imunologia , Células Dendríticas/citologia , Camundongos , Camundongos Knockout , Proteínas Quimioatraentes de Monócitos/genética , Proteínas Quimioatraentes de Monócitos/imunologia , Monócitos/citologia , Plasmócitos/citologia , Receptores de Quimiocinas/genética
10.
Immunol Rev ; 234(1): 259-67, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20193024

RESUMO

Dendritic cells (DCs) in the intestine are heterogeneous. Phenotypically different populations of conventional DCs have been identified in the intestinal lamina propria, Peyer's patches, and in the draining mesenteric lymph nodes, to which these DCs constitutively migrate. Markers used to identify these populations include major histocompatibility complex class II, CD11c, CD8 alpha, CD11b, and CD103. Extensive studies in rats, summarized here, which involved collection of migrating DCs by thoracic duct cannulation after mesenteric lymphadenectomy, have clearly demonstrated that the subsets of migrating intestinal lymph DCs have different functional properties. The subsets might play different roles in the induction of oral tolerance and in driving systemic immune responses after vaccination or intestinal stimulation with Toll-like receptor ligands. The use of these surgical techniques allows investigation of the functions of purified subsets of migrating DCs. However, in the rat, these studies are limited by the range of available reagents and are difficult to compare with data from other species in this fast-moving field. Recent refinements have enabled the collection of migrating intestinal DCs from mice; our initial results are described here. We believe that these studies will generate exciting data and have the potential to resolve important questions about the functions of migrating intestinal DC subsets.


Assuntos
Movimento Celular , Células Dendríticas/imunologia , Mucosa Intestinal/imunologia , Linfonodos/imunologia , Nódulos Linfáticos Agregados/imunologia , Animais , Antígenos CD/imunologia , Antígeno CD11b/imunologia , Antígenos CD8/imunologia , Humanos , Cadeias alfa de Integrinas/imunologia , Mucosa Intestinal/citologia , Linfa/citologia , Linfa/imunologia , Linfonodos/citologia , Camundongos , Nódulos Linfáticos Agregados/citologia , Fenótipo , Ratos , Transdução de Sinais
11.
Cell Rep ; 43(5): 114153, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38687643

RESUMO

Gut-draining mesenteric and celiac lymph nodes (mLNs and celLNs) critically contribute to peripheral tolerance toward food and microbial antigens by supporting the de novo induction of regulatory T cells (Tregs). These tolerogenic properties of mLNs and celLNs are stably imprinted within stromal cells (SCs) by microbial signals and vitamin A (VA), respectively. Here, we report that a single, transient gastrointestinal infection in the neonatal, but not adult, period durably abrogates the efficient Treg-inducing capacity of celLNs by altering the subset composition and gene expression profile of celLNSCs. These cells carry information about the early-life pathogen encounter until adulthood and durably instruct migratory dendritic cells entering the celLN with reduced tolerogenic properties. Mechanistically, transiently reduced VA levels cause long-lasting celLN functional impairment, which can be rescued by early-life treatment with VA. Together, our data highlight the therapeutic potential of VA to prevent sequelae post gastrointestinal infections in infants.


Assuntos
Linfonodos , Linfócitos T Reguladores , Vitamina A , Animais , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/efeitos dos fármacos , Vitamina A/farmacologia , Vitamina A/uso terapêutico , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Camundongos , Animais Recém-Nascidos , Tolerância Imunológica/efeitos dos fármacos , Células Dendríticas/imunologia , Camundongos Endogâmicos C57BL , Feminino
12.
Blood ; 118(23): 6220-9, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21979941

RESUMO

Lymphatic endothelial cells are important for efficient flow of antigen-bearing fluid and antigen-presenting cells (APCs) from peripheral sites to lymph nodes (LNs). APC movement to LNs is dependent on the constitutive chemokine receptor CCR7, although how conflicting inflammatory and constitutive chemokine cues are integrated at lymphatic surfaces during this process is not understood. Here we reveal a previously unrecognized aspect of the regulation of this process. The D6 chemokine-scavenging receptor, which is expressed on lymphatic endothelial cells (LECs), maintains lymphatic surfaces free of inflammatory CC-chemokines and minimizes interaction of inflammatory leukocytes with these surfaces. D6 does not alter the level of CCR7 ligands on LECs, thus ensuring selective presentation of homeostatic chemokines for interaction with CCR7(+) APCs. Accordingly, in D6-deficient mice, inflammatory CC-chemokine adherence to LECs results in inappropriate perilymphatic accumulation of inflammatory leukocytes at peripheral inflamed sites and draining LNs. This results in lymphatic congestion and impaired movement of APCs, and fluid, from inflamed sites to LNs. We propose that D6, by suppressing inflammatory chemokine binding to lymphatic surfaces, and thereby preventing inappropriate inflammatory leukocyte adherence, is a key regulator of lymphatic function and a novel, and indispensable, contributor to the integration of innate and adaptive immune responses.


Assuntos
Líquidos Corporais/imunologia , Movimento Celular/imunologia , Células Endoteliais/imunologia , Linfonodos/imunologia , Receptores de Quimiocinas/imunologia , Imunidade Adaptativa/imunologia , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Quimiocina CCL2/imunologia , Quimiocina CCL2/metabolismo , Quimiocina CCL5/imunologia , Quimiocina CCL5/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Imunidade Inata/imunologia , Leucócitos/citologia , Leucócitos/imunologia , Linfa/imunologia , Linfa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Receptores CCR2/imunologia , Receptores CCR2/metabolismo , Receptores CCR7/imunologia , Receptores CCR7/metabolismo , Receptores de Quimiocinas/genética
13.
Arthritis Rheum ; 64(10): 3199-209, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22674414

RESUMO

OBJECTIVE: In rats transgenic for human HLA-B27 and ß(2) -microglobulin (B27-transgenic rats), colitis and peripheral inflammation develop spontaneously. Therefore, B27-transgenic rats provide a model of spondylarthritis. Because inflammation in these rats requires CD4+ T lymphocytes and involves intestinal pathology, we hypothesized that dendritic cells (DCs) that migrate from the intestine and control CD4+ T cell differentiation would be aberrant in B27-transgenic rats. METHODS: Migrating intestinal lymph DCs were collected via thoracic duct cannulation from B27-transgenic and control (HLA-B7-transgenic or nontransgenic) rats. The phenotypes of these DCs and of mesenteric lymph node DCs were assessed by flow cytometry. The ability of DCs to differentiate from bone marrow precursors in vitro was also assessed. RESULTS: Lymph DCs showed increased activation and, strikingly, lacked the specific DC population that is important for maintaining tolerance to self-antigens. This population of DCs was also depleted from the mesenteric lymph nodes of B27-transgenic rats. Furthermore, in vitro culture of DCs from bone marrow precursors revealed a defect in the ability of B27-transgenic rats to produce DCs of the migratory phenotype, although the DCs that were generated induced enhanced interleukin-17 (IL-17) production from naive CD4+ T cells. CONCLUSION: We describe 2 different mechanisms by which HLA-B27 may contribute to inflammatory disease: increased apoptotic death of B27-transgenic DCs that normally function to maintain immunologic tolerance and enhanced IL-17 production from CD4+ T cells stimulated by the surviving B27-transgenic DCs.


Assuntos
Movimento Celular/genética , Células Dendríticas/imunologia , Antígeno HLA-B27/genética , Intestinos/imunologia , Espondilartrite/genética , Animais , Autoantígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Movimento Celular/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Modelos Animais de Doenças , Antígeno HLA-B27/metabolismo , Tolerância Imunológica/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-17/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/patologia , Ratos , Ratos Transgênicos , Espondilartrite/imunologia , Espondilartrite/patologia
14.
Sci Rep ; 13(1): 1509, 2023 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-36707699

RESUMO

Macrophages are traditionally considered antigen-presenting cells. However, their ability to present antigen and the factors regulating macrophage MHCII expression are poorly understood. Here, we demonstrate that MHCII expression on murine intestinal macrophages is differentially controlled by their residence in the small intestine (SI) or the colon, their ontogeny and the gut microbiota. Monocyte-derived macrophages are uniformly MHCIIhi, independently of the tissue of residence, microbial status or the age of the mouse, suggesting a common monocyte differentiation pathway. In contrast, MHCII expression on long-lived, prenatally-derived Tim4+ macrophages is low after birth but significantly increases at weaning in both SI and colon. Furthermore, MHCII expression on colonic Tim4+, but not monocyte-derived macrophages, is dependent on recognition of microbial stimuli, as MHCII expression is significantly downregulated in germ-free, antibiotic-treated and MyD88 deficient mice. To address the function of MHCII presentation by intestinal macrophages we established two models of macrophage-specific MHCII deficiency. We observed a significant reduction in the overall frequency and number of tissue-resident, but not newly arrived, SI CD4+ T cells in the absence of macrophage-expressed MHCII. Our data suggest that macrophage MHCII provides signals regulating gut CD4+ T cell maintenance with different requirements in the SI and colon.


Assuntos
Macrófagos , Microbiota , Animais , Camundongos , Colo , Homeostase , Intestino Delgado/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Antígenos de Histocompatibilidade Classe II/metabolismo
15.
Nat Rev Gastroenterol Hepatol ; 20(7): 447-461, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37085614

RESUMO

The gut and the liver are characterized by mutual interactions between both organs, the microbiome, diet and other environmental factors. The sum of these interactions is conceptualized as the gut-liver axis. In this Review we discuss the gut-liver axis, concentrating on the barriers formed by the enterohepatic tissues to restrict gut-derived microorganisms, microbial stimuli and dietary constituents. In addition, we discuss the establishment of barriers in the gut and liver during development and their cooperative function in the adult host. We detail the interplay between microbial and dietary metabolites, the intestinal epithelium, vascular endothelium, the immune system and the various host soluble factors, and how this interplay establishes a homeostatic balance in the healthy gut and liver. Finally, we highlight how this balance is disrupted in diseases of the gut and liver, outline the existing therapeutics and describe the cutting-edge discoveries that could lead to the development of novel treatment approaches.


Assuntos
Microbioma Gastrointestinal , Microbiota , Humanos , Fígado , Homeostase
16.
Discov Immunol ; 2(1): kyad018, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38567056

RESUMO

Cross-talk between dendritic cells (DCs) and the intestinal epithelium is important in the decision to mount a protective immune response to a pathogen or to regulate potentially damaging responses to food antigens and the microbiota. Failures in this decision-making process contribute to the development of intestinal inflammation, making the molecular signals that pass between DCs and intestinal epithelial cells potential therapeutic targets. Until now, in vitro models with sufficient complexity to understand these interactions have been lacking. Here, we outline the development of a co-culture model of in vitro differentiated 'gut-like' DCs with small intestinal organoids (enteroids). Sequential exposure of murine bone marrow progenitors to Flt3L, granulocyte macrophage colony-stimulating factor (GM-CSF) and all-trans-retinoic acid (RA) resulted in the generation of a distinct population of conventional DCs expressing CD11b+SIRPα+CD103+/- (cDC2) exhibiting retinaldehyde dehydrogenase (RALDH) activity. These 'gut-like' DCs extended transepithelial dendrites across the intact epithelium of enteroids. 'Gut-like' DC in co-culture with enteroids can be utilized to define how epithelial cells and cDCs communicate in the intestine under a variety of different physiological conditions, including exposure to different nutrients, natural products, components of the microbiota, or pathogens. Surprisingly, we found that co-culture with enteroids resulted in a loss of RALDH activity in 'gut-like' DCs. Continued provision of GM-CSF and RA during co-culture was required to oppose putative negative signals from the enteroid epithelium. Our data contribute to a growing understanding of how intestinal cDCs assess environmental conditions to ensure appropriate activation of the immune response.

17.
Sci Immunol ; 8(89): eadj5789, 2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-37874251

RESUMO

Regulatory T cells (Tregs) are present in lymphoid and nonlymphoid tissues where they restrict immune activation, prevent autoimmunity, and regulate inflammation. Tregs in nonlymphoid tissues are typically resident, whereas those in lymph nodes (LNs) are considered to recirculate. However, Tregs in LNs are not a homogenous population, and circulation kinetics of different Treg subsets are poorly characterized. Furthermore, whether Tregs can acquire memory T cell properties and persist for extended periods after their activation in LNs is unclear. Here, we used in situ labeling with a stabilized photoconvertible protein to uncover turnover rates of Tregs in LNs in vivo. We found that, whereas most Tregs in LNs recirculate, 10 to 20% are memory-like resident cells that remain in their respective LNs for weeks to months. Single-cell RNA sequencing revealed that LN-resident cells are a functionally and ontogenetically heterogeneous population and share the same core residency gene signature with conventional CD4+ and CD8+ T cells. Resident cells in LNs did not actively proliferate and did not require continuous T cell receptor (TCR) signaling for their residency. However, resident and circulating Tregs had distinct TCR repertoires, and each LN contained exclusive clonal subpopulations of resident Tregs. Our results demonstrate that, similar to conventional T cells, Tregs can form resident memory-like populations in LNs after adaptive immune responses. Specific and local suppression of immune responses by resident Tregs in draining LNs might provide previously unidentified therapeutic opportunities for the treatment of local chronic inflammatory conditions.


Assuntos
Linfócitos T CD8-Positivos , Linfócitos T Reguladores , Linfonodos , Transdução de Sinais , Receptores de Antígenos de Linfócitos T/metabolismo
18.
Mucosal Immunol ; 15(1): 40-50, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34465895

RESUMO

The intestine is constantly exposed to foreign antigens, which are mostly innocuous but can sometimes be harmful. Therefore, the intestinal immune system has the delicate task of maintaining immune tolerance to harmless food antigens while inducing tailored immune responses to pathogens and regulating but tolerating the microbiota. Intestinal dendritic cells (DCs) play a central role in these functions as sentinel cells able to prime and polarize the T cell responses. DCs are deployed throughout the intestinal mucosa but with local specializations along the gut length and between the diffuse effector sites of the gut lamina propria (LP) and the well-organized immune inductive sites comprising isolated lymphoid follicles (ILFs), Peyer's patches (PPs), and other species-specific gut-associated lymphoid tissues (GALTs). Understanding the specificities of each intestinal DC subset, how environmental factors influence DC functions, and how these can be modulated is key to harnessing the therapeutic potential of mucosal adaptive immune responses, whether by enhancing the efficacy of mucosal vaccines or by increasing tolerogenic responses in inflammatory disorders. In this review, we summarize recent findings related to intestinal DCs in steady state and upon inflammation, with a special focus on their functional specializations, highly dependent on their microenvironment.


Assuntos
Células Dendríticas/imunologia , Imunomodulação/imunologia , Mucosa Intestinal/imunologia , Intestinos/imunologia , Tecido Linfoide/imunologia , Animais , Humanos , Imunidade nas Mucosas
19.
Cell Host Microbe ; 30(11): 1630-1645.e25, 2022 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-36208631

RESUMO

Microbiome research needs comprehensive repositories of cultured bacteria from the intestine of mammalian hosts. We expanded the mouse intestinal bacterial collection (www.dsmz.de/miBC) to 212 strains, all publicly available and taxonomically described. This includes strain-level diversity, small-sized bacteria, and previously undescribed taxa (one family, 10 genera, and 39 species). This collection enabled metagenome-educated prediction of synthetic communities (SYNs) that capture key functional differences between microbiomes, notably identifying communities associated with either resistance or susceptibility to DSS-induced colitis. Additionally, nine species were used to amend the Oligo-Mouse Microbiota (OMM)12 model, yielding the OMM19.1 model. The added strains compensated for phenotype differences between OMM12 and specific pathogen-free mice, including body composition and immune cells in the intestine and associated lymphoid tissues. Ready-to-use OMM stocks are available for future studies. In conclusion, this work improves our knowledge of gut microbiota diversity in mice and enables functional studies via the modular use of isolates.


Assuntos
Microbioma Gastrointestinal , Microbiota , Camundongos , Animais , Microbioma Gastrointestinal/genética , Bactérias , Metagenoma , Intestinos , Modelos Animais de Doenças , Mamíferos/genética
20.
J Immunol ; 182(4): 2405-15, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19201895

RESUMO

Dendritic cells (DCs) are crucial to intestinal immune regulation because of their roles in inducing protective immunity against pathogens while maintaining tolerance to commensal bacteria. Nonetheless, relatively little is known about intestinal DC responsiveness to innate immune stimuli via TLRs. We have previously shown that DCs migrating from the rat intestine in lymph (iLDCs) are hyporesponsive to LPS stimulation, thus possibly preventing harmful immune responses being induced to commensal flora. In this study, to understand how iLDC function is regulated by innate immune stimuli, we have characterized the expression and function of TLRs in iLDCs isolated from the thoracic duct lymph of mesenteric lymphadenectomized rats and compared these with DCs grown from bone marrow in the presence of Flt3 ligand. We show that iLDCs express mRNAs for all TLRs, but express significantly less TLR4 mRNA than bone marrow-derived DCs. Functionally, iLDCs could be activated by TLR agonists representing intestinal pathogen-associated molecular patterns, with the important exception of the TLR4 agonist LPS. Furthermore, we show that DCs in the intestinal wall interact directly with noninvasive bacteria (Bacillus subtilis spores), leading to an increase in the output of activated iLDCs into lymph, and that DCs containing spores are activated selectively. These data highlight a functional difference between TLR4 and other TLRs. As iLDCs can respond to TLR stimulation in vitro, there must be other mechanisms that prevent their activation by commensal bacteria under steady-state conditions.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/imunologia , Intestinos/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Bacillus subtilis/imunologia , Células da Medula Óssea/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunidade nas Mucosas/imunologia , Intestinos/citologia , Lipopolissacarídeos/imunologia , RNA Mensageiro/análise , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Toll-Like/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA