Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Microbiol ; 9(3): 614-630, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38429422

ABSTRACT

Microbial transformation of bile acids affects intestinal immune homoeostasis but its impact on inflammatory pathologies remains largely unknown. Using a mouse model of graft-versus-host disease (GVHD), we found that T cell-driven inflammation decreased the abundance of microbiome-encoded bile salt hydrolase (BSH) genes and reduced the levels of unconjugated and microbe-derived bile acids. Several microbe-derived bile acids attenuated farnesoid X receptor (FXR) activation, suggesting that loss of these metabolites during inflammation may increase FXR activity and exacerbate the course of disease. Indeed, mortality increased with pharmacological activation of FXR and decreased with its genetic ablation in donor T cells during mouse GVHD. Furthermore, patients with GVHD after allogeneic hematopoietic cell transplantation showed similar loss of BSH and the associated reduction in unconjugated and microbe-derived bile acids. In addition, the FXR antagonist ursodeoxycholic acid reduced the proliferation of human T cells and was associated with a lower risk of GVHD-related mortality in patients. We propose that dysbiosis and loss of microbe-derived bile acids during inflammation may be an important mechanism to amplify T cell-mediated diseases.


Subject(s)
Graft vs Host Disease , T-Lymphocytes , Humans , Intestines , Inflammation , Bile Acids and Salts
2.
Immunity ; 55(7): 1173-1184.e7, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35700740

ABSTRACT

Regulatory T (Treg) cells expressing the transcription factor Foxp3 are an essential suppressive T cell lineage of dual origin: Foxp3 induction in thymocytes and mature CD4+ T cells gives rise to thymic (tTreg) and peripheral (pTreg) Treg cells, respectively. While tTreg cells suppress autoimmunity, pTreg cells enforce tolerance to food and commensal microbiota. However, the role of Foxp3 in pTreg cells and the mechanisms supporting their differentiation remain poorly understood. Here, we used genetic tracing to identify microbiota-induced pTreg cells and found that many of their distinguishing features were Foxp3 independent. Lineage-committed, microbiota-dependent pTreg-like cells persisted in the colon in the absence of Foxp3. While Foxp3 was critical for the suppression of a Th17 cell program, colitis, and mastocytosis, pTreg cells suppressed colonic effector T cell expansion in a Foxp3-independent manner. Thus, Foxp3 and the tolerogenic signals that precede and promote its expression independently confer distinct facets of pTreg functionality.


Subject(s)
Forkhead Transcription Factors , T-Lymphocytes, Regulatory , Forkhead Transcription Factors/metabolism , Immune Tolerance , Th17 Cells/metabolism , Thymocytes/metabolism
3.
Cell Metab ; 33(5): 851-852, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33951469

ABSTRACT

Glycolysis supports effector T cell function but is detrimental to the immunosuppressive activity of regulatory T cells. In a recent issue of Nature, two papers address a role for glucose and lactate availability within the tumor microenvironment for the balance of pro- and anti-tumoral effects of T cells and the efficacy of neoadjuvant cancer immunotherapy.


Subject(s)
Neoplasms , T-Lymphocytes, Regulatory , Humans , Immunotherapy , Neoplasms/therapy , Sugars , Tumor Microenvironment
4.
J Exp Med ; 218(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33373442

ABSTRACT

T cells increase cholesterol biosynthesis upon activation to generate substrates for cellular growth and proliferation. The ubiquitously expressed liver X receptor ß (LXRß) encoded by the Nr1h2 gene is a critical regulator of cholesterol homeostasis in mammalian cells; however, its cell-intrinsic role in T cell biology remains poorly understood. We report that ablation of LXRß in T cells leads to spontaneous T cell activation and T lymphocytopenia. Unexpectedly, analysis of mixed bone marrow chimeric mice revealed a cell-autonomous survival defect that reduced the fitness of LXRß-deficient effector T cells, suggesting that the heightened immune activation in mice harboring LXRß-deficient T cells was due to impaired regulatory T (T reg) cell functionality. Indeed, we found that single-copy deletion of Nr1h2 in T reg cells disrupted activated T reg cell metabolism and fitness and resulted in early-onset fatal autoimmune disease. Our study demonstrated an indispensable requirement for T reg cell-intrinsic LXRß function in immune homeostasis and provides a basis for immunological therapies through targeting of this receptor.


Subject(s)
Autoimmune Diseases/immunology , Homeostasis/immunology , Liver X Receptors/physiology , Lymphocyte Activation/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytopenia, Idiopathic CD4-Positive/immunology , Animals , Autoimmune Diseases/genetics , Cells, Cultured , Cholesterol/metabolism , Female , Forkhead Transcription Factors/genetics , Homeostasis/genetics , Liver X Receptors/genetics , Male , Mice , Mice, Inbred C57BL , Radiation Chimera/immunology , Signal Transduction/genetics , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytopenia, Idiopathic CD4-Positive/genetics
5.
Proc Natl Acad Sci U S A ; 117(52): 33446-33454, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33318189

ABSTRACT

Reduced nutrient intake is a widely conserved manifestation of sickness behavior with poorly characterized effects on adaptive immune responses. During infectious challenges, naive T cells encountering their cognate antigen become activated and differentiate into highly proliferative effector T cells. Despite their evident metabolic shift upon activation, it remains unclear how effector T cells respond to changes in nutrient availability in vivo. Here, we show that spontaneous or imposed feeding reduction during infection decreases the numbers of splenic lymphocytes. Effector T cells showed cell-intrinsic responses dependent on the nuclear receptor Farnesoid X Receptor (FXR). Deletion of FXR in T cells prevented starvation-induced loss of lymphocytes and increased effector T cell fitness in nutrient-limiting conditions, but imparted greater weight loss to the host. FXR deficiency increased the contribution of glutamine and fatty acids toward respiration and enhanced cell survival under low-glucose conditions. Provision of glucose during anorexia of infection rescued effector T cells, suggesting that this sugar is a limiting nutrient for activated lymphocytes and that alternative fuel usage may affect cell survival in starved animals. Altogether, we identified a mechanism by which the host scales immune responses according to food intake, featuring FXR as a T cell-intrinsic sensor.


Subject(s)
Feeding Behavior , Lymphocytic Choriomeningitis/immunology , Receptors, Cytoplasmic and Nuclear/metabolism , T-Lymphocytes/immunology , Animals , Anorexia/virology , Fasting , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Nutrients/metabolism , Spleen/pathology , Transcription, Genetic
6.
Nature ; 581(7809): 475-479, 2020 05.
Article in English | MEDLINE | ID: mdl-32461639

ABSTRACT

Intestinal health relies on the immunosuppressive activity of CD4+ regulatory T (Treg) cells1. Expression of the transcription factor Foxp3 defines this lineage, and can be induced extrathymically by dietary or commensal-derived antigens in a process assisted by a Foxp3 enhancer known as conserved non-coding sequence 1 (CNS1)2-4. Products of microbial fermentation including butyrate facilitate the generation of peripherally induced Treg (pTreg) cells5-7, indicating that metabolites shape the composition of the colonic immune cell population. In addition to dietary components, bacteria modify host-derived molecules, generating a number of biologically active substances. This is epitomized by the bacterial transformation of bile acids, which creates a complex pool of steroids8 with a range of physiological functions9. Here we screened the major species of deconjugated bile acids for their ability to potentiate the differentiation of pTreg cells. We found that the secondary bile acid 3ß-hydroxydeoxycholic acid (isoDCA) increased Foxp3 induction by acting on dendritic cells (DCs) to diminish their immunostimulatory properties. Ablating one receptor, the farnesoid X receptor, in DCs enhanced the generation of Treg cells and imposed a transcriptional profile similar to that induced by isoDCA, suggesting an interaction between this bile acid and nuclear receptor. To investigate isoDCA in vivo, we took a synthetic biology approach and designed minimal microbial consortia containing engineered Bacteroides strains. IsoDCA-producing consortia increased the number of colonic RORγt-expressing Treg cells in a CNS1-dependent manner, suggesting enhanced extrathymic differentiation.


Subject(s)
Bacteria/metabolism , Bile Acids and Salts/chemistry , Bile Acids and Salts/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Amino Acid Sequence , Animals , Bacteroides/metabolism , Colon/microbiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Fermentation , Gastrointestinal Microbiome , Male , Mice , Mice, Inbred C57BL , Microbial Consortia , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
7.
Cell Metab ; 31(1): 18-25, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31607562

ABSTRACT

Regulatory T (Treg) cells expressing the X-chromosome-encoded transcription factor Foxp3 represent a specialized immunosuppressive lineage with a well-recognized, essential function in preventing fatal autoimmunity and inflammation. Recent studies revealed that Treg cells can also exert systemic effects on metabolism and partake in tissue repair, suggesting a dual role for these cells in serving and protecting tissues. Here, we review multiple means by which Treg cells support tissue function and organismal homeostasis.


Subject(s)
Adaptive Immunity , Autoimmunity , Forkhead Transcription Factors/metabolism , Inflammation/physiopathology , T-Lymphocytes, Regulatory/immunology , Animals , Homeostasis/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Regeneration/immunology , Regeneration/physiology
8.
Immunity ; 48(6): 1245-1257.e9, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29858010

ABSTRACT

The mammalian gut microbiota provides essential metabolites to the host and promotes the differentiation and accumulation of extrathymically generated regulatory T (pTreg) cells. To explore the impact of these cells on intestinal microbial communities, we assessed the composition of the microbiota in pTreg cell-deficient and -sufficient mice. pTreg cell deficiency led to heightened type 2 immune responses triggered by microbial exposure, which disrupted the niche of border-dwelling bacteria early during colonization. Moreover, impaired pTreg cell generation led to pervasive changes in metabolite profiles, altered features of the intestinal epithelium, and reduced body weight in the presence of commensal microbes. Absence of a single species of bacteria depleted in pTreg cell-deficient animals, Mucispirillum schaedleri, partially accounted for the sequelae of pTreg cell deficiency. These observations suggest that pTreg cells modulate the metabolic function of the intestinal microbiota by restraining immune defense mechanisms that may disrupt a particular bacterial niche.


Subject(s)
Gastrointestinal Microbiome/immunology , Host Microbial Interactions/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Mice
9.
Shock ; 45(4): 393-403, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26618986

ABSTRACT

Dysregulation of the inflammatory response against infection contributes to mortality in sepsis. Inflammation provides critical host defense, but it can cause tissue damage, multiple organ failure, and death. Because the nuclear transcription factor peroxisome proliferator-activated receptor γ (PPARγ) exhibits therapeutic potential, we characterized the role of PPARγ in sepsis. We analyzed severity of clinical signs, survival rates, cytokine production, leukocyte influx, and bacterial clearance in a cecal ligation and puncture (CLP) model of sepsis in Swiss mice. The PPARγ agonist rosiglitazone treatment improved clinical status and mortality, while increasing IL-10 production and decreasing TNF-α and IL-6 levels, and peritoneal neutrophil accumulation 24 h after CLP. We noted increased bacterial killing in rosiglitazone treated mice, correlated with increased generation of reactive oxygen species. Polymorphonuclear leukocytes (PMN) incubated with LPS or Escherichia coli and rosiglitazone increased peritoneal neutrophil extracellular trap (NET)-mediated bacterial killing, an effect reversed by the PPARγ antagonist (GW 9662) treatment. Rosiglitazone also enhanced the release of histones by PMN, a surrogate marker of NET formation, effect abolished by GW 9662. Rosiglitazone modulated the inflammatory response and increased bacterial clearance through PPARγ activation and NET formation, combining immunomodulatory and host-dependent anti-bacterial effects and, therefore, warrants further study as a potential therapeutic agent in sepsis.


Subject(s)
Escherichia coli/immunology , Extracellular Traps/immunology , Neutrophils/immunology , PPAR gamma/agonists , Sepsis/drug therapy , Signal Transduction/drug effects , Thiazolidinediones/pharmacology , Anilides/pharmacology , Animals , Disease Models, Animal , Male , Mice , PPAR gamma/immunology , Rosiglitazone , Sepsis/immunology , Sepsis/microbiology , Sepsis/pathology , Signal Transduction/immunology
10.
Nature ; 504(7480): 451-5, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24226773

ABSTRACT

Intestinal microbes provide multicellular hosts with nutrients and confer resistance to infection. The delicate balance between pro- and anti-inflammatory mechanisms, essential for gut immune homeostasis, is affected by the composition of the commensal microbial community. Regulatory T cells (Treg cells) expressing transcription factor Foxp3 have a key role in limiting inflammatory responses in the intestine. Although specific members of the commensal microbial community have been found to potentiate the generation of anti-inflammatory Treg or pro-inflammatory T helper 17 (TH17) cells, the molecular cues driving this process remain elusive. Considering the vital metabolic function afforded by commensal microorganisms, we reasoned that their metabolic by-products are sensed by cells of the immune system and affect the balance between pro- and anti-inflammatory cells. We tested this hypothesis by exploring the effect of microbial metabolites on the generation of anti-inflammatory Treg cells. We found that in mice a short-chain fatty acid (SCFA), butyrate, produced by commensal microorganisms during starch fermentation, facilitated extrathymic generation of Treg cells. A boost in Treg-cell numbers after provision of butyrate was due to potentiation of extrathymic differentiation of Treg cells, as the observed phenomenon was dependent on intronic enhancer CNS1 (conserved non-coding sequence 1), essential for extrathymic but dispensable for thymic Treg-cell differentiation. In addition to butyrate, de novo Treg-cell generation in the periphery was potentiated by propionate, another SCFA of microbial origin capable of histone deacetylase (HDAC) inhibition, but not acetate, which lacks this HDAC-inhibitory activity. Our results suggest that bacterial metabolites mediate communication between the commensal microbiota and the immune system, affecting the balance between pro- and anti-inflammatory mechanisms.


Subject(s)
Butyrates/metabolism , Cell Differentiation , Intestinal Mucosa/metabolism , Intestines/microbiology , Symbiosis , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Acetylation , Animals , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enhancer Elements, Genetic/genetics , Fermentation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Histone Deacetylases/metabolism , Inflammation Mediators/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestines/cytology , Intestines/immunology , Introns/genetics , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Starch/metabolism , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...