Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Science ; 382(6666): eadg2253, 2023 10 06.
Article in English | MEDLINE | ID: mdl-37797010

ABSTRACT

Disruption of cellular activities by pathogen virulence factors can trigger innate immune responses. Interferon-γ (IFN-γ)-inducible antimicrobial factors, such as the guanylate binding proteins (GBPs), promote cell-intrinsic defense by attacking intracellular pathogens and by inducing programmed cell death. Working in human macrophages, we discovered that GBP1 expression in the absence of IFN-γ killed the cells and induced Golgi fragmentation. IFN-γ exposure improved macrophage survival through the activity of the kinase PIM1. PIM1 phosphorylated GBP1, leading to its sequestration by 14-3-3σ, which thereby prevented GBP1 membrane association. During Toxoplasma gondii infection, the virulence protein TgIST interfered with IFN-γ signaling and depleted PIM1, thereby increasing GBP1 activity. Although infected cells can restrain pathogens in a GBP1-dependent manner, this mechanism can protect uninfected bystander cells. Thus, PIM1 can provide a bait for pathogen virulence factors, guarding the integrity of IFN-γ signaling.


Subject(s)
GTP-Binding Proteins , Host-Pathogen Interactions , Immunity, Innate , Interferon-gamma , Proto-Oncogene Proteins c-pim-1 , Toxoplasma , Toxoplasmosis , Humans , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Interferon-gamma/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Toxoplasmosis/immunology , Virulence Factors/metabolism , Macrophages/immunology , 14-3-3 Proteins/metabolism , Host-Pathogen Interactions/immunology
2.
Immunology ; 166(3): 299-309, 2022 07.
Article in English | MEDLINE | ID: mdl-35322416

ABSTRACT

In CD4+ T helper cells, the active form of vitamin D3 , 1,25-dihydroxyvitamin D3 (1,25D) suppresses production of inflammatory cytokines, including interferon-gamma (IFN-γ), but the mechanisms for this are not yet fully defined. In innate immune cells, response to 1,25D has been linked to metabolic reprogramming. It is unclear whether 1,25D has similar effects on CD4+ T cells, although it is known that antigen stimulation of these cells promotes an anabolic metabolic phenotype, characterized by high rates of aerobic glycolysis to support clonal expansion and effector cytokine expression. Here, we performed in-depth analysis of metabolic capacity and pathway usage, employing extracellular flux and stable isotope-based tracing approaches, in CD4+ T cells treated with 1,25D. We report that 1,25D significantly decreases rates of aerobic glycolysis in activated CD4+ T cells, whilst exerting a lesser effect on mitochondrial glucose oxidation. This is associated with transcriptional repression of Myc, but not repression of mTOR activity under these conditions. Consistent with the modest effect of 1,25D on mitochondrial activity, it also did not impact CD4+ T-cell mitochondrial mass or membrane potential. Finally, we demonstrate that inhibition of aerobic glycolysis by 1,25D substantially contributes to its immune-regulatory capacity in CD4+ T cells, since the suppression of IFN-γ expression was significantly blunted in the absence of aerobic glycolysis. 1,25-Dihydroxyvitamin D3 (1,25D) suppresses the production of inflammatory cytokines such as interferon-gamma (IFN-γ) by CD4+ T cells, but the underpinning mechanisms are not yet fully defined. Here, we identify that 1,25D inhibits aerobic glycolysis in activated CD4+ T cells, associated with decreased c-Myc expression. This mechanism appears to substantially contribute to the suppression of IFN-γ by 1,25D, since this is significantly blunted in the absence of aerobic glycolysis.


Subject(s)
Calcitriol , Interferon-gamma , Calcitriol/metabolism , Calcitriol/pharmacology , Glycolysis , Interferon-gamma/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Vitamin D
3.
Life Sci Alliance ; 5(6)2022 06.
Article in English | MEDLINE | ID: mdl-35256513

ABSTRACT

Inflammasomes are cytosolic innate immune sensors of pathogen infection and cellular damage that induce caspase-1-mediated inflammation upon activation. Although inflammation is protective, uncontrolled excessive inflammation can cause inflammatory diseases and can be detrimental, such as in coronavirus disease (COVID-19). However, the underlying mechanisms that control inflammasome activation are incompletely understood. Here we report that the leucine-rich repeat (LRR) protein ribonuclease inhibitor (RNH1), which shares homology with LRRs of NLRP (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing) proteins, attenuates inflammasome activation. Deletion of RNH1 in macrophages increases interleukin (IL)-1ß production and caspase-1 activation in response to inflammasome stimulation. Mechanistically, RNH1 decreases pro-IL-1ß expression and induces proteasome-mediated caspase-1 degradation. Corroborating this, mouse models of monosodium urate (MSU)-induced peritonitis and lipopolysaccharide (LPS)-induced endotoxemia, which are dependent on caspase-1, respectively, show increased neutrophil infiltration and lethality in Rnh1 -/- mice compared with wild-type mice. Furthermore, RNH1 protein levels were negatively related with disease severity and inflammation in hospitalized COVID-19 patients. We propose that RNH1 is a new inflammasome regulator with relevance to COVID-19 severity.


Subject(s)
COVID-19/pathology , Carrier Proteins/metabolism , Inflammasomes/metabolism , Leucine-Rich Repeat Proteins/metabolism , Animals , COVID-19/immunology , Caspase 1/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Patient Acuity , Proteasome Endopeptidase Complex/metabolism
4.
Discov Immunol ; 1(1): kyac009, 2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36704407

ABSTRACT

In lymphocytes, Nr4a gene expression is specifically regulated by antigen receptor signalling, making them ideal targets for use as distal T cell receptor (TCR) reporters. Nr4a3-Timer of cell kinetics and activity (Tocky) mice are a ground-breaking tool to report TCR-driven Nr4a3 expression using Fluorescent Timer protein (FT). FT undergoes a time-dependent shift in its emission spectrum following translation, allowing for the temporal reporting of transcriptional events. Our recent work suggested that Nr4a1/Nur77 may be a more sensitive gene to distal TCR signals compared to Nr4a3, so we, therefore, generated Nur77-Timer-rapidly-expressed-in-lymphocytes (Tempo) mice that express FT under the regulation of Nur77. We validated the ability of Nur77-Tempo mice to report TCR and B cell receptor signals and investigated the signals regulating Nur77-FT expression. We found that Nur77-FT was sensitive to low-strength TCR signals, and its brightness was graded in response to TCR signal strength. Nur77-FT detected positive selection signals in the thymus, and analysis of FT expression revealed that positive selection signals are often persistent in nature, with most thymic Treg expressing FT Blue. We found that active TCR signals in the spleen are low frequency, but CD69+ lymphoid T cells are enriched for FT Blue+ Red+ T cells, suggesting frequent TCR signalling. In non-lymphoid tissue, we saw a dissociation of FT protein from CD69 expression, indicating that tissue residency is not associated with tonic TCR signals. Nur77-Tempo mice, therefore, combine the temporal dynamics from the Tocky innovation with increased sensitivity of Nr4a1 to lower TCR signal strengths.

5.
Biochem Soc Trans ; 49(6): 2601-2610, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34854889

ABSTRACT

The gut relies on the complex interaction between epithelial, stromal and immune cells to maintain gut health in the face of food particles and pathogens. Innate sensing by the intestinal epithelium is critical for maintaining epithelial barrier function and also orchestrating mucosal immune responses. Numerous innate pattern recognition receptors (PRRs) are involved in such sensing. In recent years, several Nucleotide-binding-domain and Leucine-rich repeat-containing receptors (NLRs) have been found to partake in pathogen or damage sensing while also being implicated in gut pathologies, such as colitis and colorectal cancer (CRC). Here, we discuss the current literature focusing on NLR family apoptosis inhibitory proteins (NAIPs) and other NLRs that have non-inflammasome roles in the gut. The mechanisms behind NLR-mediated protection often converges on similar signalling pathways, such as STAT3, MAPK and NFκB. Further understanding of how these NLRs contribute to the maintenance of gut homeostasis will be important for understanding gut pathologies and developing new therapies.


Subject(s)
Inflammasomes/metabolism , Intestinal Mucosa/metabolism , NLR Proteins/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Animals , Colorectal Neoplasms/metabolism , Humans
6.
JCI Insight ; 6(23)2021 10 28.
Article in English | MEDLINE | ID: mdl-34710062

ABSTRACT

Bacterial cancer therapy (BCT) shows great promise for treatment of solid tumors, yet basic mechanisms of bacterial-induced tumor suppression remain undefined. Attenuated strains of Salmonella enterica serovar Typhimurium (STm) have commonly been used in mouse models of BCT in xenograft and orthotopic transplant cancer models. We aimed to better understand the tumor epithelium-targeted mechanisms of BCT by using autochthonous mouse models of intestinal cancer and tumor organoid cultures to assess the effectiveness and consequences of oral treatment with aromatase A-deficient STm (STmΔaroA). STmΔaroA delivered by oral gavage significantly reduced tumor burden and tumor load in both a colitis-associated colorectal cancer (CAC) model and in a spontaneous Apcmin/+ intestinal cancer model. STmΔaroA colonization of tumors caused alterations in transcription of mRNAs associated with tumor stemness, epithelial-mesenchymal transition, and cell cycle. Metabolomic analysis of tumors demonstrated alteration in the metabolic environment of STmΔaroA-treated tumors, suggesting that STmΔaroA imposes metabolic competition on the tumor. Use of tumor organoid cultures in vitro recapitulated effects seen on tumor stemness, mesenchymal markers, and altered metabolome. Furthermore, live STmΔaroA was required, demonstrating active mechanisms including metabolite usage. We have demonstrated that oral BCT is efficacious in autochthonous intestinal cancer models, that BCT imposes metabolic competition, and that BCT has direct effects on the tumor epithelium affecting tumor stem cells.


Subject(s)
Biological Therapy , Colorectal Neoplasms/therapy , Salmonella typhimurium/physiology , Administration, Oral , Animals , Aromatase/metabolism , Disease Models, Animal , Epithelium , Mice , Organoids , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics
7.
Immunity ; 54(11): 2481-2496.e6, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34534438

ABSTRACT

How T cell receptor (TCR) signal strength modulates T cell function and to what extent this is modified by immune checkpoint blockade (ICB) are key questions in immunology. Using Nr4a3-Tocky mice, we characterized early quantitative and qualitative changes that occur in CD4+ T cells in relation to TCR signaling strength. We captured how dose- and time-dependent programming of distinct co-inhibitory receptors rapidly recalibrates T cell activation thresholds and visualized the immediate effects of ICB on T cell re-activation. Our findings reveal that anti-PD1 immunotherapy leads to an increased TCR signal strength. We defined a strong TCR signal metric of five genes upregulated by anti-PD1 in T cells (TCR.strong), which was superior to a canonical T cell activation gene signature in stratifying melanoma patient outcomes to anti-PD1 therapy. Our study therefore reveals how analysis of TCR signal strength-and its manipulation-can provide powerful metrics for monitoring outcomes to immunotherapy.


Subject(s)
Antigens/immunology , Immune Checkpoint Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Gene Expression Regulation , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Proteins/genetics , Lymphocyte Activation , Melanoma/drug therapy , Melanoma/etiology , Melanoma/metabolism , Melanoma/pathology , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , T-Lymphocytes/drug effects
9.
J Clin Invest ; 128(4): 1597-1614, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29408805

ABSTRACT

Ribosomal proteins (RP) regulate specific gene expression by selectively translating subsets of mRNAs. Indeed, in Diamond-Blackfan anemia and 5q- syndrome, mutations in RP genes lead to a specific defect in erythroid gene translation and cause anemia. Little is known about the molecular mechanisms of selective mRNA translation and involvement of ribosomal-associated factors in this process. Ribonuclease inhibitor 1 (RNH1) is a ubiquitously expressed protein that binds to and inhibits pancreatic-type ribonucleases. Here, we report that RNH1 binds to ribosomes and regulates erythropoiesis by controlling translation of the erythroid transcription factor GATA1. Rnh1-deficient mice die between embryonic days E8.5 and E10 due to impaired production of mature erythroid cells from progenitor cells. In Rnh1-deficient embryos, mRNA levels of Gata1 are normal, but GATA1 protein levels are decreased. At the molecular level, we found that RNH1 binds to the 40S subunit of ribosomes and facilitates polysome formation on Gata1 mRNA to confer transcript-specific translation. Further, RNH1 knockdown in human CD34+ progenitor cells decreased erythroid differentiation without affecting myelopoiesis. Our results reveal an unsuspected role for RNH1 in the control of GATA1 mRNA translation and erythropoiesis.


Subject(s)
Embryo, Mammalian/metabolism , Erythropoiesis , GATA1 Transcription Factor/biosynthesis , Hematopoietic Stem Cells/metabolism , Protein Biosynthesis , Proteins/metabolism , Animals , Embryo, Mammalian/cytology , GATA1 Transcription Factor/genetics , Hematopoietic Stem Cells/cytology , Humans , K562 Cells , Mice , Mice, Knockout , Proteins/genetics , Ribosome Subunits, Large/genetics , Ribosome Subunits, Large/metabolism
10.
Trends Immunol ; 36(8): 442-50, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26166583

ABSTRACT

While the functional importance of inflammasomes in blood-derived cell types is well established, it remains poorly understood how inflammasomes in nonhematopoietic cells contribute to mucosal immunity. Recent studies have revealed functional roles of inflammasomes - particularly NAIP/NLRC4, NLRP6, and noncanonical caspase-4 (caspase-11) - within epithelial cells of the gut in mucosal immune defense, inflammation, and tumorigenesis. Here, we review and discuss these findings in the broader context of tissue compartment-specific mucosal immunity. We propose several models whereby activities of the intestinal epithelial inflammasomes converge on mechanisms to remove compromised epithelial cells, maintain host-microbiota mutualism, and communicate with immune cells of the underlying lamina propria.


Subject(s)
Inflammasomes/immunology , Intestinal Mucosa/immunology , Animals , Humans , Immunity, Mucosal
11.
Arthritis Rheumatol ; 67(6): 1646-56, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25914377

ABSTRACT

OBJECTIVE: Host-microbial interactions are central in health and disease. Monosodium urate monohydrate (MSU) crystals cause gout by activating the NLRP3 inflammasome, leading to interleukin-1ß (IL-1ß) production and neutrophil recruitment. This study was undertaken to investigate the relevance of gut microbiota, acetate, and the metabolite-sensing receptor GPR43 in regulating inflammation in a murine model of gout. METHODS: Gout was induced by the injection of MSU crystals into the knee joints of mice. Macrophages from the various animals were stimulated to determine inflammasome activation and production of reactive oxygen species (ROS). RESULTS: Injection of MSU crystals caused joint inflammation, as seen by neutrophil influx, hypernociception, and production of IL-1ß and CXCL1. These parameters were greatly decreased in germ-free mice, mice treated with antibiotics, and GPR-43-deficient mice. Recolonization or administration of acetate to germ-free mice restored inflammation in response to injection of MSU crystals. In vitro, macrophages produced ROS and assembled the inflammasome when stimulated with MSU. Macrophages from germ-free animals produced little ROS, and there was little inflammasome assembly. Similar results were observed in macrophages from GPR-43-deficient mice. Treatment of germ-free mice with acetate restored in vitro responsiveness of macrophages to MSU crystals. CONCLUSION: In the absence of microbiota, there is decreased production of short-chain fatty acids that are necessary for adequate inflammasome assembly and IL-1ß production in a manner that is at least partially dependent on GPR43. These results clearly show that the commensal microbiota shapes the host's ability to respond to an inflammasome-dependent acute inflammatory stimulus outside the gut.


Subject(s)
Fatty Acids, Volatile/metabolism , Gout/genetics , Inflammasomes/immunology , Macrophages/immunology , Microbiota/immunology , Reactive Oxygen Species/immunology , Receptors, G-Protein-Coupled/genetics , Animals , Cell Movement , Chemokine CXCL1/immunology , Disease Models, Animal , Gout/immunology , Hyperalgesia , Interleukin-1beta/immunology , Intestines/microbiology , Mice , Neutrophils , Nociceptive Pain , Receptors, G-Protein-Coupled/immunology , Sodium Acetate , Stifle , Uric Acid
12.
J Exp Med ; 212(3): 369-83, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25732303

ABSTRACT

NLR family apoptosis inhibitory proteins (NAIPs) belong to both the Nod-like receptor (NLR) and the inhibitor of apoptosis (IAP) families. NAIPs are known to form an inflammasome with NLRC4, but other in vivo functions remain unexplored. Using mice deficient for all NAIP paralogs (Naip1-6(Δ/Δ)), we show that NAIPs are key regulators of colorectal tumorigenesis. Naip1-6(Δ/Δ) mice developed increased colorectal tumors, in an epithelial-intrinsic manner, in a model of colitis-associated cancer. Increased tumorigenesis, however, was not driven by an exacerbated inflammatory response. Instead, Naip1-6(Δ/Δ) mice were protected from severe colitis and displayed increased antiapoptotic and proliferation-related gene expression. Naip1-6(Δ/Δ) mice also displayed increased tumorigenesis in an inflammation-independent model of colorectal cancer. Moreover, Naip1-6(Δ/Δ) mice, but not Nlrc4-null mice, displayed hyper-activation of STAT3 and failed to activate p53 18 h after carcinogen exposure. This suggests that NAIPs protect against tumor initiation in the colon by promoting the removal of carcinogen-elicited epithelium, likely in a NLRC4 inflammasome-independent manner. Collectively, we demonstrate a novel epithelial-intrinsic function of NAIPs in protecting the colonic epithelium against tumorigenesis.


Subject(s)
Colitis/pathology , Colonic Neoplasms/pathology , Neuronal Apoptosis-Inhibitory Protein/metabolism , Animals , Colitis/genetics , Colitis/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Inflammasomes/genetics , Inflammasomes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neuronal Apoptosis-Inhibitory Protein/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
13.
Cell Host Microbe ; 16(2): 237-248, 2014 Aug 13.
Article in English | MEDLINE | ID: mdl-25121751

ABSTRACT

The gut mucosal epithelium separates the host from the microbiota, but enteropathogens such as Salmonella Typhimurium (S.Tm) can invade and breach this barrier. Defenses against such acute insults remain incompletely understood. Using a murine model of Salmonella enterocolitis, we analyzed mechanisms limiting pathogen loads in the epithelium during early infection. Although the epithelium-invading S.Tm replicate initially, this intraepithelial replicative niche is restricted by expulsion of infected enterocytes into the lumen. This mechanism is compromised if inflammasome components (NAIP1-6, NLRC4, caspase-1/-11) are deleted, or ablated specifically in the epithelium, resulting in ∼100-fold higher intraepithelial loads and accelerated lymph node colonization. Interestingly, the cytokines downstream of inflammasome activation, interleukin (IL)-1α/ß and IL-18, appear dispensable for epithelial restriction of early infection. These data establish the role of an epithelium-intrinsic inflammasome, which drives expulsion of infected cells to restrict the pathogen's intraepithelial proliferation. This may represent a general defense mechanism against mucosal infections.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Calcium-Binding Proteins/physiology , Enterocytes/microbiology , Neuronal Apoptosis-Inhibitory Protein/physiology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Animals , Cecum/microbiology , Cecum/pathology , Enterocytes/immunology , Host-Pathogen Interactions , Inflammasomes , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice, Inbred C57BL , Mice, Knockout
14.
PLoS Pathog ; 9(8): e1003531, 2013.
Article in English | MEDLINE | ID: mdl-23935505

ABSTRACT

Approximately 2 billion people currently suffer from intestinal helminth infections, which are typically chronic in nature and result in growth retardation, vitamin A deficiency, anemia and poor cognitive function. Such chronicity results from co-evolution between helminths and their mammalian hosts; however, the molecular mechanisms by which these organisms avert immune rejection are not clear. We have found that the natural murine helminth, Heligmosomoides polygyrus bakeri (Hp) elicits the secretion of IL-1ß in vivo and in vitro and that this cytokine is critical for shaping a mucosal environment suited to helminth chronicity. Indeed in mice deficient for IL-1ß (IL-1ß(-/-)), or treated with the soluble IL-1ßR antagonist, Anakinra, helminth infection results in enhanced type 2 immunity and accelerated parasite expulsion. IL-1ß acts to decrease production of IL-25 and IL-33 at early time points following infection and parasite rejection was determined to require IL-25. Taken together, these data indicate that Hp promotes the release of host-derived IL-1ß that suppresses the release of innate cytokines, resulting in suboptimal type 2 immunity and allowing pathogen chronicity.


Subject(s)
Immunity, Innate , Immunity, Mucosal , Interleukin-1beta/immunology , Interleukins/immunology , Nematospiroides dubius/immunology , Strongylida Infections/immunology , Animals , Antirheumatic Agents/pharmacology , Chronic Disease , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/genetics , Interleukin-33 , Interleukins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Strongylida Infections/genetics , Strongylida Infections/pathology , Th2 Cells/immunology
15.
Immunol Rev ; 245(1): 164-76, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22168419

ABSTRACT

Certain autoimmune diseases as well as asthma have increased in recent decades, particularly in developed countries. The hygiene hypothesis has been the prevailing model to account for this increase; however, epidemiology studies also support the contribution of diet and obesity to inflammatory diseases. Diet affects the composition of the gut microbiota, and recent studies have identified various molecules and mechanisms that connect diet, the gut microbiota, and immune responses. Herein, we discuss the effects of microbial metabolites, such as short chain fatty acids, on epithelial integrity as well as immune cell function. We propose that dysbiosis contributes to compromised epithelial integrity and disrupted immune tolerance. In addition, dietary molecules affect the function of immune cells directly, particularly through lipid G-protein coupled receptors such as GPR43.


Subject(s)
Bacteria/immunology , Bacterial Infections/immunology , Fatty Acids/immunology , Immune System Diseases/immunology , Intestinal Mucosa/immunology , Animals , Bacteria/metabolism , Bacterial Infections/complications , Bacterial Infections/microbiology , Diet , Dietary Supplements , Fatty Acids/metabolism , Humans , Immune System Diseases/etiology , Immune System Diseases/microbiology , Immune Tolerance , Inflammation/immunology , Inflammation/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Receptors, G-Protein-Coupled/metabolism
16.
Semin Immunol ; 23(2): 139-45, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21292499

ABSTRACT

The gut microbiota has recently been recognized for its role in immune regulation, and changes in gut microbiota may be the basis for an increased incidence of autoimmune diseases and asthma in developed countries. Beneficial microbes produce factors that are distributed systemically, and therefore can influence peripheral inflammatory responses. Such symbiosis factors are important for the control and resolution of inflammation and autoimmune diseases. Here we discuss immune regulation by recently identified symbiosis factors and how certain environmental factors favor their production and influence the composition of the gut microflora.


Subject(s)
Autoimmunity , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Animals , Autoimmune Diseases/immunology , Humans , Inflammatory Bowel Diseases/microbiology , Symbiosis
17.
Nat Immunol ; 12(1): 5-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21169997

ABSTRACT

The fields of immunology, microbiology, nutrition and metabolism are rapidly converging. Here we expand on a diet-microbiota model as the basis for the greater incidence of asthma and autoimmunity in developed countries.


Subject(s)
Diet , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Lipid Metabolism/immunology , Metagenome/immunology , Animals , Clinical Trials as Topic , Genetic Predisposition to Disease , Humans , Immunity, Mucosal/genetics , Immunomodulation , Inflammation , Mice , Models, Immunological , Signal Transduction/genetics , Signal Transduction/immunology
18.
Nature ; 461(7268): 1282-6, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19865172

ABSTRACT

The immune system responds to pathogens by a variety of pattern recognition molecules such as the Toll-like receptors (TLRs), which promote recognition of dangerous foreign pathogens. However, recent evidence indicates that normal intestinal microbiota might also positively influence immune responses, and protect against the development of inflammatory diseases. One of these elements may be short-chain fatty acids (SCFAs), which are produced by fermentation of dietary fibre by intestinal microbiota. A feature of human ulcerative colitis and other colitic diseases is a change in 'healthy' microbiota such as Bifidobacterium and Bacteriodes, and a concurrent reduction in SCFAs. Moreover, increased intake of fermentable dietary fibre, or SCFAs, seems to be clinically beneficial in the treatment of colitis. SCFAs bind the G-protein-coupled receptor 43 (GPR43, also known as FFAR2), and here we show that SCFA-GPR43 interactions profoundly affect inflammatory responses. Stimulation of GPR43 by SCFAs was necessary for the normal resolution of certain inflammatory responses, because GPR43-deficient (Gpr43(-/-)) mice showed exacerbated or unresolving inflammation in models of colitis, arthritis and asthma. This seemed to relate to increased production of inflammatory mediators by Gpr43(-/-) immune cells, and increased immune cell recruitment. Germ-free mice, which are devoid of bacteria and express little or no SCFAs, showed a similar dysregulation of certain inflammatory responses. GPR43 binding of SCFAs potentially provides a molecular link between diet, gastrointestinal bacterial metabolism, and immune and inflammatory responses.


Subject(s)
Chemotactic Factors/metabolism , Inflammation/metabolism , Inflammation/microbiology , Intestines/microbiology , Receptors, G-Protein-Coupled/metabolism , Acetates/therapeutic use , Animals , Arthritis/metabolism , Cells, Cultured , Colitis/drug therapy , Colitis/metabolism , Colitis/microbiology , Fatty Acids, Volatile/metabolism , Germ-Free Life , Humans , Inflammation/drug therapy , Metagenome , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Oligonucleotide Array Sequence Analysis , Protein Array Analysis , Receptors, G-Protein-Coupled/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...