Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Cell ; 187(4): 897-913.e18, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38280374

ABSTRACT

Canonically, the complement system is known for its rapid response to remove microbes in the bloodstream. However, relatively little is known about a functioning complement system on intestinal mucosal surfaces. Herein, we report the local synthesis of complement component 3 (C3) in the gut, primarily by stromal cells. C3 is expressed upon commensal colonization and is regulated by the composition of the microbiota in healthy humans and mice, leading to an individual host's specific luminal C3 levels. The absence of membrane attack complex (MAC) components in the gut ensures that C3 deposition does not result in the lysis of commensals. Pathogen infection triggers the immune system to recruit neutrophils to the infection site for pathogen clearance. Basal C3 levels directly correlate with protection against enteric infection. Our study reveals the gut complement system as an innate immune mechanism acting as a vigilant sentinel that combats pathogens and spares commensals.


Subject(s)
Complement C3 , Intestinal Mucosa , Microbiota , Animals , Humans , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Neutrophils , Complement C3/metabolism , Stromal Cells/metabolism
2.
Immunity ; 57(4): 876-889.e11, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38479384

ABSTRACT

Concentrations of the secondary bile acid, deoxycholic acid (DCA), are aberrantly elevated in colorectal cancer (CRC) patients, but the consequences remain poorly understood. Here, we screened a library of gut microbiota-derived metabolites and identified DCA as a negative regulator for CD8+ T cell effector function. Mechanistically, DCA suppressed CD8+ T cell responses by targeting plasma membrane Ca2+ ATPase (PMCA) to inhibit Ca2+-nuclear factor of activated T cells (NFAT)2 signaling. In CRC patients, CD8+ T cell effector function negatively correlated with both DCA concentration and expression of a bacterial DCA biosynthetic gene. Bacteria harboring DCA biosynthetic genes suppressed CD8+ T cells effector function and promoted tumor growth in mice. This effect was abolished by disrupting bile acid metabolism via bile acid chelation, genetic ablation of bacterial DCA biosynthetic pathway, or specific bacteriophage. Our study demonstrated causation between microbial DCA metabolism and anti-tumor CD8+ T cell response in CRC, suggesting potential directions for anti-tumor therapy.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Humans , Mice , Animals , Bile Acids and Salts , Deoxycholic Acid/pharmacology , CD8-Positive T-Lymphocytes
3.
Immunity ; 56(12): 2773-2789.e8, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37992711

ABSTRACT

Although the gut microbiota can influence central nervous system (CNS) autoimmune diseases, the contribution of the intestinal epithelium to CNS autoimmunity is less clear. Here, we showed that intestinal epithelial dopamine D2 receptors (IEC DRD2) promoted sex-specific disease progression in an animal model of multiple sclerosis. Female mice lacking Drd2 selectively in intestinal epithelial cells showed a blunted inflammatory response in the CNS and reduced disease progression. In contrast, overexpression or activation of IEC DRD2 by phenylethylamine administration exacerbated disease severity. This was accompanied by altered lysozyme expression and gut microbiota composition, including reduced abundance of Lactobacillus species. Furthermore, treatment with N2-acetyl-L-lysine, a metabolite derived from Lactobacillus, suppressed microglial activation and neurodegeneration. Taken together, our study indicates that IEC DRD2 hyperactivity impacts gut microbial abundances and increases susceptibility to CNS autoimmune diseases in a female-biased manner, opening up future avenues for sex-specific interventions of CNS autoimmune diseases.


Subject(s)
Autoimmune Diseases of the Nervous System , Multiple Sclerosis , Male , Female , Mice , Animals , Multiple Sclerosis/metabolism , Disease Models, Animal , Signal Transduction , Disease Progression , Receptors, Dopamine
4.
Nature ; 619(7971): 837-843, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37380774

ABSTRACT

The human gut microbiome constantly converts natural products derived from the host and diet into numerous bioactive metabolites1-3. Dietary fats are essential micronutrients that undergo lipolysis to release free fatty acids (FAs) for absorption in the small intestine4. Gut commensal bacteria modify some unsaturated FAs-for example, linoleic acid (LA)-into various intestinal FA isomers that regulate host metabolism and have anticarcinogenic properties5. However, little is known about how this diet-microorganism FA isomerization network affects the mucosal immune system of the host. Here we report that both dietary factors and microbial factors influence the level of gut LA isomers (conjugated LAs (CLAs)) and that CLAs in turn modulate a distinct population of CD4+ intraepithelial lymphocytes (IELs) that express CD8αα in the small intestine. Genetic abolition of FA isomerization pathways in individual gut symbionts significantly decreases the number of CD4+CD8αα+ IELs in gnotobiotic mice. Restoration of CLAs increases CD4+CD8αα+ IEL levels in the presence of the transcription factor hepatocyte nuclear factor 4γ (HNF4γ). Mechanistically, HNF4γ facilitates CD4+CD8αα+ IEL development by modulating interleukin-18 signalling. In mice, specific deletion of HNF4γ in T cells leads to early mortality from infection by intestinal pathogens. Our data reveal a new role for bacterial FA metabolic pathways in the control of host intraepithelial immunological homeostasis by modulating the relative number of CD4+ T cells that were CD4+CD8αα+.


Subject(s)
Fatty Acids , Gastrointestinal Microbiome , Intraepithelial Lymphocytes , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Isomerism , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lipolysis , Linoleic Acid/metabolism , Immunity, Mucosal
5.
Immunity ; 50(3): 692-706.e7, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30824326

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a severe form of lung fibrosis with a high mortality rate. However, the etiology of IPF remains unknown. Here, we report that alterations in lung microbiota critically promote pulmonary fibrosis pathogenesis. We found that lung microbiota was dysregulated, and the dysregulated microbiota in turn induced production of interleukin-17B (IL-17B) during bleomycin-induced mouse lung fibrosis. Either lung-microbiota depletion or IL-17B deficiency ameliorated the disease progression. IL-17B cooperated with tumor necrosis factor-α to induce expression of neutrophil-recruiting genes and T helper 17 (Th17)-cell-promoting genes. Three pulmonary commensal microbes, which belong to the genera Bacteroides and Prevotella, were identified to promote fibrotic pathogenesis through IL-17R signaling. We further defined that the outer membrane vesicles (OMVs) that were derived from the identified commensal microbes induced IL-17B production through Toll-like receptor-Myd88 adaptor signaling. Together our data demonstrate that specific pulmonary symbiotic commensals can promote lung fibrosis by regulating a profibrotic inflammatory cytokine network.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/microbiology , Interleukin-17/metabolism , Lung/metabolism , Lung/microbiology , Microbiota/physiology , Animals , Bacteroides/metabolism , Cytokines/metabolism , Disease Models, Animal , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Neutrophils/metabolism , Prevotella/metabolism , Signal Transduction/physiology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Nature ; 577(7791): 543-548, 2020 01.
Article in English | MEDLINE | ID: mdl-31915378

ABSTRACT

Although maternal antibodies protect newborn babies from infection1,2, little is known about how protective antibodies are induced without prior pathogen exposure. Here we show that neonatal mice that lack the capacity to produce IgG are protected from infection with the enteric pathogen enterotoxigenic Escherichia coli by maternal natural IgG antibodies against the maternal microbiota when antibodies are delivered either across the placenta or through breast milk. By challenging pups that were fostered by either maternal antibody-sufficient or antibody-deficient dams, we found that IgG derived from breast milk was crucial for protection against mucosal disease induced by enterotoxigenic E. coli. IgG also provides protection against systemic infection by E. coli. Pups used the neonatal Fc receptor to transfer IgG from milk into serum. The maternal commensal microbiota can induce antibodies that recognize antigens expressed by enterotoxigenic E. coli and other Enterobacteriaceae species. Induction of maternal antibodies against a commensal Pantoea species confers protection against enterotoxigenic E. coli in pups. This role of the microbiota in eliciting protective antibodies to a specific neonatal pathogen represents an important host defence mechanism against infection in neonates.


Subject(s)
Antibodies/immunology , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Immunity, Maternally-Acquired/immunology , Infant, Newborn/immunology , Microbiota/immunology , Milk, Human/immunology , Animals , Antibodies/blood , Antibodies/metabolism , Breast Feeding , Cross Reactions/immunology , Escherichia coli Infections/microbiology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Male , Mice , Mothers , Pantoea/immunology , Receptors, Fc/immunology , Receptors, Fc/metabolism , Symbiosis/immunology
7.
Nature ; 577(7790): 410-415, 2020 01.
Article in English | MEDLINE | ID: mdl-31875848

ABSTRACT

The metabolic pathways encoded by the human gut microbiome constantly interact with host gene products through numerous bioactive molecules1. Primary bile acids (BAs) are synthesized within hepatocytes and released into the duodenum to facilitate absorption of lipids or fat-soluble vitamins2. Some BAs (approximately 5%) escape into the colon, where gut commensal bacteria convert them into various intestinal BAs2 that are important hormones that regulate host cholesterol metabolism and energy balance via several nuclear receptors and/or G-protein-coupled receptors3,4. These receptors have pivotal roles in shaping host innate immune responses1,5. However, the effect of this host-microorganism biliary network on the adaptive immune system remains poorly characterized. Here we report that both dietary and microbial factors influence the composition of the gut BA pool and modulate an important population of colonic FOXP3+ regulatory T (Treg) cells expressing the transcription factor RORγ. Genetic abolition of BA metabolic pathways in individual gut symbionts significantly decreases this Treg cell population. Restoration of the intestinal BA pool increases colonic RORγ+ Treg cell counts and ameliorates host susceptibility to inflammatory colitis via BA nuclear receptors. Thus, a pan-genomic biliary network interaction between hosts and their bacterial symbionts can control host immunological homeostasis via the resulting metabolites.


Subject(s)
Bile Acids and Salts/metabolism , Gastrointestinal Microbiome , Homeostasis , Intestines/immunology , Intestines/microbiology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bile Acids and Salts/chemistry , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
8.
Immunity ; 43(3): 488-501, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26320657

ABSTRACT

The intestinal epithelial barrier plays a critical role in the mucosal immunity. However, it remains largely unknown how the epithelial barrier is maintained after damage. Here we show that growth factor FGF2 synergized with interleukin-17 (IL-17) to induce genes for repairing of damaged epithelium. FGF2 or IL-17 deficiency resulted in impaired epithelial proliferation, increased pro-inflammatory microbiota outgrowth, and consequently worse pathology in a DSS-induced colitis model. The dysregulated microbiota in the model induced transforming growth factor beta 1 (TGFß1) expression, which in turn induced FGF2 expression mainly in regulatory T cells. Act1, an essential adaptor in IL-17 signaling, suppressed FGF2-induced ERK activation through binding to adaptor molecule GRB2 to interfere with its association with guanine nucleotide exchange factor SOS1. Act1 preferentially bound to IL-17 receptor complex, releasing its suppressive effect on FGF2 signaling. Thus, microbiota-driven FGF2 and IL-17 cooperate to repair the damaged intestinal epithelium through Act1-mediated direct signaling cross-talk.


Subject(s)
Fibroblast Growth Factor 2/immunology , Interleukin-17/immunology , Intestines/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression Profiling/methods , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Interleukin-17/genetics , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestines/microbiology , Intestines/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microbiota/genetics , Microbiota/immunology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Wound Healing/immunology
9.
EMBO J ; 38(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30770344

ABSTRACT

T helper 17 (Th17)-cell differentiation triggered by interleukin-6 (IL-6) via STAT3 activation promotes inflammation in inflammatory bowel disease (IBD) patients. However, leukemia inhibitory factor (LIF), an IL-6 family cytokine, restricts inflammation by blocking Th17-cell differentiation via an unknown mechanism. Here, we report that microbiota dysregulation promotes LIF secretion by intestinal epithelial cells (IECs) in a mouse colitis model. LIF greatly activates STAT4 phosphorylation on multiple SPXX elements within the C-terminal transcription regulation domain. STAT4 and STAT3 act reciprocally on both canonical cis-inducible elements (SIEs) and noncanonical "AGG" elements at different loci. In lamina propria lymphocytes (LPLs), STAT4 activation by LIF blocks STAT3-dependent Il17a/Il17f promoter activation, whereas in IECs, LIF bypasses the extraordinarily low level of STAT4 to induce YAP gene expression via STAT3 activation. In addition, we found that the administration of LIF is sufficient to restore microbiome homeostasis. Thus, LIF effectively inhibits Th17 accumulation and promotes repair of damaged intestinal epithelium in inflamed colon, serves as a potential therapy for IBD.


Subject(s)
Colitis/prevention & control , Gene Expression Regulation/drug effects , Inflammation/prevention & control , Intestinal Mucosa/drug effects , Leukemia Inhibitory Factor/pharmacology , STAT3 Transcription Factor/metabolism , STAT4 Transcription Factor/physiology , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/immunology , Inflammation/chemically induced , Inflammation/immunology , Interleukin-17/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , STAT3 Transcription Factor/genetics , Signal Transduction , Th17 Cells/immunology
10.
Nat Immunol ; 12(12): 1151-8, 2011 Oct 12.
Article in English | MEDLINE | ID: mdl-21993849

ABSTRACT

Interleukin 17 receptor E (IL-17RE) is an orphan receptor of the IL-17 receptor family. Here we show that IL-17RE is a receptor specific to IL-17C and has an essential role in host mucosal defense against infection. IL-17C activated downstream signaling through IL-17RE-IL-17RA complex for the induction of genes encoding antibacterial peptides as well as proinflammatory molecules. IL-17C was upregulated in colon epithelial cells during infection with Citrobacter rodentium and acted in synergy with IL-22 to induce the expression of antibacterial peptides in colon epithelial cells. Loss of IL-17C-mediated signaling in IL-17RE-deficient mice led to lower expression of genes encoding antibacterial molecules, greater bacterial burden and early mortality during infection. Together our data identify IL-17RE as a receptor of IL-17C that regulates early innate immunity to intestinal pathogens.


Subject(s)
Enterobacteriaceae Infections/immunology , Immunity, Mucosal/immunology , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Receptors, Interleukin-17/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Cell Line , Citrobacter rodentium , Colon/immunology , Colon/metabolism , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/microbiology , Gene Expression Regulation , HEK293 Cells , HT29 Cells , Humans , Immunity, Mucosal/genetics , Interleukins/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-17/genetics , Signal Transduction , Interleukin-22
11.
Immunity ; 40(1): 140-52, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24412611

ABSTRACT

Although the microbiota has been shown to drive production of interleukin-17A (IL-17A) from T helper 17 cells to promote cell proliferation and tumor growth in colorectal cancer, the molecular mechanisms for microbiota-mediated regulation of tumorigenesis are largely unknown. Here, we found that the innate-like cytokine IL-17C was upregulated in human colorectal cancers and in mouse intestinal tumor models. Alterations in the microbiota drove IL-17C upregulation specifically in intestinal epithelial cells (IECs) through Toll-like receptor (TLR)-MyD88-dependent signaling during intestinal tumorigenesis. Microbiota-driven IL-17C induced Bcl-2 and Bcl-xL expression in IECs in an autocrine manner to promote cell survival and tumorigenesis in both chemically induced and spontaneous intestinal tumor models. Thus, IL-17C promotes cancer development by increasing IEC survival, and the microbiota can mediate cancer pathogenesis through regulation of IL-17C.


Subject(s)
Carcinogenesis/immunology , Colonic Neoplasms/immunology , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Microbiota/immunology , Animals , Autocrine Communication , Cell Survival , Cells, Cultured , Colonic Neoplasms/microbiology , Disease Models, Animal , Humans , Interleukin-17/genetics , Intestinal Mucosa/microbiology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Up-Regulation , bcl-X Protein/genetics , bcl-X Protein/metabolism
12.
Int J Mol Sci ; 22(22)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34830307

ABSTRACT

Melatonin, a widely known indoleamine molecule that mediates various animal and plant physiological processes, is formed from N-acetyl serotonin via N-acetylserotonin methyltransferase (ASMT). ASMT is an enzyme that catalyzes melatonin synthesis in plants in the rate-determining step and is homologous to hydroxyindole-O-methyltransferase (HIOMT) melatonin synthase in animals. To date, little is known about the effect of HIOMT on salinity in apple plants. Here, we explored the melatonin physiological function in the salinity condition response by heterologous expressing the homologous human HIOMT gene in apple plants. We discovered that the expression of melatonin-related gene (MdASMT) in apple plants was induced by salinity. Most notably, compared with the wild type, three transgenic lines indicated higher melatonin levels, and the heterologous expression of HIOMT enhanced the expression of melatonin synthesis genes. The transgenic lines showed reduced salt damage symptoms, lower relative electrolyte leakage, and less total chlorophyll loss from leaves under salt stress. Meanwhile, through enhanced activity of antioxidant enzymes, transgenic lines decreased the reactive oxygen species accumulation, downregulated the expression of the abscisic acid synthesis gene (MdNCED3), accordingly reducing the accumulation of abscisic acid under salt stress. Both mechanisms regulated morphological changes in the stomata synergistically, thereby mitigating damage to the plants' photosynthetic ability. In addition, transgenic plants also effectively stabilized their ion balance, raised the expression of salt stress-related genes, as well as alleviated osmotic stress through changes in amino acid metabolism. In summary, heterologous expression of HIOMT improved the adaptation of apple leaves to salt stress, primarily by increasing melatonin concentration, maintaining a high photosynthetic capacity, reducing reactive oxygen species accumulation, and maintaining normal ion homeostasis.


Subject(s)
Acetylserotonin O-Methyltransferase/genetics , Gene Expression Regulation, Plant , Genes, Plant , Malus/genetics , Melatonin/genetics , Abscisic Acid/metabolism , Amino Acids/metabolism , Chlorophyll/metabolism , Homeostasis/genetics , Ions/metabolism , Malus/growth & development , Malus/metabolism , Melatonin/metabolism , Osmotic Pressure , Photosynthesis/genetics , Plant Development/genetics , Plant Stomata/genetics , Plant Stomata/growth & development , Plant Stomata/metabolism , Plants, Genetically Modified , Reactive Oxygen Species/metabolism , Salinity , Salt Tolerance/genetics , Signal Transduction/genetics
13.
Immunology ; 159(2): 156-166, 2020 02.
Article in English | MEDLINE | ID: mdl-31631335

ABSTRACT

Host-microbiota interaction plays fundamental roles in the homeostasis of mucosal immunity. Dysbiosis of intestinal microbiota has been demonstrated to participate in various immune responses and many multifactorial diseases. Study of intestinal microbiota has moved beyond the consequences of dysbiosis to the causal microbiota associated with diseases. However, studies of pulmonary microbiota and its dysbiosis are still in their infancy. Improvement of culture-dependent and -independent techniques has facilitated our understanding of lung microbiota that not only exists in healthy lung tissue but also exerts great impact on immune responses under both physiological and pathological conditions. In this review, we summarize recent progresses of lung microbiota dysbiosis and its impact on the local immune system that determines the balance of tolerance and inflammation. We discuss the causal roles of pulmonary dysbiosis under disease settings, and propose that the interaction between lung microbiota and host is critical for establishing the immune homeostasis in lung.


Subject(s)
Dysbiosis , Lung/microbiology , Microbiota , Pneumonia/microbiology , Adaptive Immunity , Animals , Host-Pathogen Interactions , Humans , Immunity, Innate , Lung/immunology , Lung/metabolism , Pneumonia/immunology , Pneumonia/metabolism
14.
Adv Exp Med Biol ; 841: 99-151, 2014.
Article in English | MEDLINE | ID: mdl-25261206

ABSTRACT

CD4(+) T helper cells are classical but constantly reinterpreted T-cell subset, playing critical roles in a diverse range of inflammatory responses or diseases. Depending on the cytokines they release and the immune responses they mediate, CD4(+) T cells are classically divided into two major cell populations: Th1 and Th2 cells. However, recent studies challenged this Th1/Th2 paradigm by discovering several T-helper cell subsets with specific differentiation program and functions, including Th17 cells, Treg cells, and Tfh cells. In this chapter, we summarize the current understanding and recent progresses on the Th17 lineage differentiation and its effector impacts on variety of inflammatory responses or disease pathogenesis.


Subject(s)
Cell Differentiation , Inflammation/immunology , Th17 Cells/physiology , Animals , Autoimmune Diseases/immunology , Cytokines/physiology , Humans , Neoplasms/immunology , T-Lymphocytes, Regulatory/physiology , TOR Serine-Threonine Kinases/physiology , Th17 Cells/cytology , Transcription Factors/physiology
15.
J Clin Invest ; 134(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38299588

ABSTRACT

Aberrant alternative splicing (AS) events have been identified in a variety of cancers. Although somatic mutations of splicing factors and dysregulation of RNA-binding proteins (RBPs) have been linked to AS and tumor malignancy, it remains unclear how upstream mechanisms contribute to cancer development via alternative gene splicing. In this issue of the JCI, Wenrui Zhang and colleagues identified the role of asparagine endopeptidase (AEP), an intracellular cysteine endopeptidase, in promoting solid tumor-associated RNA splicing. The authors demonstrated that tumor environmental factors such as oxygen and nutrient deprivation induce the activity of AEP in a HIF1A-dependent manner. The activated AEP, in turn, cleaves an RNA helicase DDX3X to promote its nuclear retention. The authors further showed that this DDX3X nuclear fraction engages with splicing machinery to induce AS events in several cancer cells. These findings suggest that targeting an AEP-dependent aberrant RNA splicing cascade may facilitate therapeutics for solid tumors.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , RNA Splicing , Alternative Splicing , DEAD-box RNA Helicases/genetics
16.
Gut Microbes ; 16(1): 2382336, 2024.
Article in English | MEDLINE | ID: mdl-39046079

ABSTRACT

Commensal microorganisms in the human gut produce numerous metabolites by using small molecules derived from the host or diet as precursors. Host or dietary lipid molecules are involved in energy metabolism and maintaining the structural integrity of cell membranes. Notably, gut microbes can convert these lipids into bioactive signaling molecules through their biotransformation and synthesis pathways. These microbiota-derived lipid metabolites can affect host physiology by influencing the body's immune and metabolic processes. This review aims to summarize recent advances in the microbial transformation and host immunomodulatory functions of these lipid metabolites, with a special focus on fatty acids and steroids produced by our gut microbiota.


Subject(s)
Biotransformation , Fatty Acids , Gastrointestinal Microbiome , Sterols , Humans , Fatty Acids/metabolism , Animals , Sterols/metabolism , Bacteria/metabolism , Immunomodulation , Lipid Metabolism
17.
Cell Host Microbe ; 32(6): 950-963.e8, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38788722

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by dysbiosis of the gut microbiota and dysfunction of intestinal stem cells (ISCs). However, the direct interactions between IBD microbial factors and ISCs are undescribed. Here, we identify α2A-adrenergic receptor (ADRA2A) as a highly expressed GPCR in ISCs. Through PRESTO-Tango screening, we demonstrate that tyramine, primarily produced by Enterococcus via tyrosine decarboxylase (tyrDC), serves as a microbial ligand for ADRA2A. Using an engineered tyrDC-deficient Enterococcus faecalis strain and intestinal epithelial cell-specific Adra2a knockout mice, we show that Enterococcus-derived tyramine suppresses ISC proliferation, thereby impairing epithelial regeneration and exacerbating DSS-induced colitis through ADRA2A. Importantly, blocking the axis with an ADRA2A antagonist, yohimbine, disrupts tyramine-mediated suppression on ISCs and alleviates colitis. Our findings highlight a microbial ligand-GPCR pair in ISCs, revealing a causal link between microbial regulation of ISCs and colitis exacerbation and yielding a targeted therapeutic approach to restore ISC function in colitis.


Subject(s)
Colitis , Mice, Knockout , Receptors, Adrenergic, alpha-2 , Stem Cells , Tyramine , Animals , Tyramine/metabolism , Tyramine/pharmacology , Colitis/microbiology , Colitis/chemically induced , Colitis/metabolism , Mice , Receptors, Adrenergic, alpha-2/metabolism , Stem Cells/metabolism , Humans , Mice, Inbred C57BL , Tyrosine Decarboxylase/metabolism , Enterococcus faecalis/metabolism , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Yohimbine/pharmacology , Disease Models, Animal , Enterococcus/metabolism , Intestines/microbiology , Intestines/pathology , Cell Proliferation , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/metabolism , Dextran Sulfate
18.
Cell Mol Gastroenterol Hepatol ; 18(2): 101350, 2024.
Article in English | MEDLINE | ID: mdl-38704148

ABSTRACT

BACKGROUND & AIMS: Gut bacterial sphingolipids, primarily produced by Bacteroidetes, have dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells. Patients with inflammatory bowel disease display altered sphingolipids profiles in fecal samples. However, how bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remains unclear. METHODS: We used dextran sodium sulfate (DSS)-induced colitis in mice monocolonized with Bacteroides fragilis strains expressing or lacking sphingolipids to assess the influence of bacterial sphingolipids on intestinal inflammation using transcriptional, protein, and cellular analyses. Colonic explant and organoid were used to study the function of bacterial sphingolipids. Host mucosal immune cells and cytokines were profiled and characterized using flow cytometry, enzyme-linked immunosorbent assay, and Western blot, and cytokine function in vivo was investigated by monoclonal antibody injection. RESULTS: B fragilis sphingolipids exacerbated intestinal inflammation. Mice monocolonized with B fragilis lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin (IL)-22 by ILC3. Mice colonized with B fragilis lacking sphingolipids following DSS treatment showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production. Protection against DSS colitis associated with B fragilis lacking sphingolipids was reversed on IL22 blockade. Furthermore, bacterial sphingolipids restricted epithelial IL18 production following DSS treatment and interfered with IL22 production by a subset of ILC3 cells expressing both IL18R and major histocompatibility complex class II. CONCLUSIONS: B fragilis-derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL18 expression and concomitantly suppressing the production of IL22 by ILC3 cells.


Subject(s)
Bacteroides fragilis , Colitis , Dextran Sulfate , Interleukin-22 , Interleukins , Sphingolipids , Animals , Sphingolipids/metabolism , Interleukins/metabolism , Mice , Colitis/immunology , Colitis/pathology , Colitis/chemically induced , Colitis/microbiology , Colitis/metabolism , Dextran Sulfate/toxicity , Bacteroides fragilis/immunology , Disease Models, Animal , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , STAT3 Transcription Factor/metabolism , Mice, Inbred C57BL
19.
J Ethnopharmacol ; 333: 118409, 2024 Oct 28.
Article in English | MEDLINE | ID: mdl-38823662

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: China and India have unique traditional medicine systems with vast territory and rich medical resources. Traditional medicines in China include traditional Chinese medicine, Tibetan medicine, Mongolian medicine, Uyghur medicine, Dai medicine, etc. In the third national survey of Chinese medicine resources, 12694 medicinal materials were identified. Traditional medicines in India include Ayurveda, Unani, Siddha, Homoeopathy, etc. There are 7263 medicinal materials in India. AIM OF THE STUDY: To reveal the characteristics of medicinal materials between China and India respectively, and to compare the similarities and differences in terms of properties, tastes, medicinal parts and therapeutic uses and to promote the exchange of traditional medicine between China and India and the international trade of traditional medicine industry. METHODS: The information of medicinal materials between China and India was extracted from The Chinese Traditional Medicine Resource Records and Pharmacopoeia of the People's Republic of China, as well as from 71 Indian herbal monographs. The information of each medicinal material, such as types, families, genera, properties, distribution, medicinal parts, efficacy, therapeutic uses, dosage form and dosage, was recorded in Excel for statistical analysis and visual comparison. RESULTS: A total of 12694 medicinal materials in China and 5362 medicinal materials in India were identified. The medicinal materials were mostly distributed in Southwest China and northern India. Plants were the main sources of medicinal materials. The common medicinal parts in China were whole medicinal materials, roots and rhizomes, and India used more renewable fruits, seeds and leaves. They are commonly used in the treatment of digestive system diseases. There were 1048 medicinal materials used by both China and India, which were distributed in 188 families and 685 genera. The Chinese and Indian pharmacopoeias had a total of 80 species of medicinal materials used by both China and India. CONCLUSIONS: The characteristics of medicinal materials between China and India were somewhat different, which was conducive to provide a reference basis for traditional medicine in China or India to increase the medicinal parts and indications when using a certain medicinal material, as well as to expand the source of medicine and introduce new resources. However, there were certain similarities and shared medicinal materials, which can tap the potential of bilateral trade of medicinal materials between China and India, so as to promote the medical cultural exchange and economic and trade cooperation between the two countries.


Subject(s)
Data Mining , Plants, Medicinal , India , China , Plants, Medicinal/chemistry , Humans , Data Mining/methods , Medicine, Chinese Traditional/methods , Medicine, Traditional/methods , Phytotherapy/methods
20.
Cytokine ; 62(2): 175-82, 2013 May.
Article in English | MEDLINE | ID: mdl-23557798

ABSTRACT

Interleukin-17 (IL-17), the signature cytokine produced by T helper 17 (Th17) cells, plays pivotal roles in host defense responses against microbial invasion, as well as in the pathogenesis of autoimmune diseases and allergic syndromes. IL-17 activates several downstream signaling pathways including NF-κB, MAPKs and C/EBPs to induce gene expression of antibacterial peptides, proinflammatory chemokines and cytokines and matrix metalloproteinases (MMPs). IL-17 can also stabilize mRNAs of genes induced by TNFα. Although the physiological and pathological functions of IL-17 have been studied for many years, the landscape of its signaling transduction has not been described until recently. The cytosolic adaptor molecule Act1 (also known as CIKS) is considered as the master mediator of IL-17 signaling. In this review, we will summarize recent progress on activation and regulation of IL-17 mediated signal transduction, especially on Act1 mediated regulation of the signaling.


Subject(s)
Interleukin-17/metabolism , Receptors, Interleukin-17/metabolism , Th17 Cells/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autoimmune Diseases/immunology , CCAAT-Enhancer-Binding Proteins/metabolism , Humans , Interleukin-17/immunology , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Receptors, Interleukin-17/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL