Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 8120, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38097562

ABSTRACT

Pulmonary fibrosis (PF), a condition characterized by inflammation and collagen deposition in the alveolar interstitium, causes dyspnea and fatal outcomes. Although the bleomycin-induced PF mouse model has improved our understanding of exogenous factor-induced fibrosis, the mechanism governing endogenous factor-induced fibrosis remains unknown. Here, we find that Ifngr1-/-Rag2-/- mice, which lack the critical suppression factor for group 2 innate lymphoid cells (ILC2), develop PF spontaneously. The onset phase of fibrosis includes ILC2 subpopulations with a high Il1rl1 (IL-33 receptor) expression, and fibrosis does not develop in ILC-deficient or IL-33-deficient mice. Although ILC2s are normally localized near bronchioles and blood vessels, ILC2s are increased in fibrotic areas along with IL-33 positive fibroblasts during fibrosis. Co-culture analysis shows that activated-ILC2s directly induce collagen production from fibroblasts. Furthermore, increased IL1RL1 and decreased IFNGR1 expressions are confirmed in ILC2s from individuals with idiopathic PF, highlighting the applicability of Ifngr1-/-Rag2-/- mice as a mouse model for fibrosis research.


Subject(s)
Pulmonary Fibrosis , Animals , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Immunity, Innate , Interleukin-33/genetics , Lymphocytes , Fibrosis , Collagen , Lung/pathology , Mice, Inbred C57BL , Interleukin-1 Receptor-Like 1 Protein
2.
J Exp Med ; 220(8)2023 08 07.
Article in English | MEDLINE | ID: mdl-37163450

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) expressing IL-5 and IL-13 are localized at various mucosal tissues and play critical roles in the induction of type 2 inflammation, response to helminth infection, and tissue repair. Here, we reveal a unique ILC2 subset in the mouse intestine that constitutively expresses IL-4 together with GATA3, ST2, KLRG1, IL-17RB, and IL-5. In this subset, IL-4 expression is regulated by mechanisms similar to but distinct from those observed in T cells and is partly affected by IL-25 signaling. Although the absence of the microbiota had marginal effects, feeding mice with a vitamin B1-deficient diet compromised the number of intestinal IL-4+ ILC2s. The decrease in the number of IL-4+ ILC2s caused by the vitamin B1 deficiency was accompanied by a reduction in IL-25-producing tuft cells. Our findings reveal that dietary vitamin B1 plays a critical role in maintaining interaction between tuft cells and IL-4+ ILC2s, a previously uncharacterized immune cell population that may contribute to maintaining intestinal homeostasis.


Subject(s)
Diet , Intestinal Mucosa , Thiamine , Animals , Mice , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Thiamine/metabolism , Specific Pathogen-Free Organisms , Mice, Inbred C57BL , Interleukin-4/metabolism , Gastrointestinal Microbiome , Organoids/cytology , Organoids/immunology , Trinitrobenzenesulfonic Acid
3.
Nat Med ; 22(6): 679-84, 2016 06.
Article in English | MEDLINE | ID: mdl-27111280

ABSTRACT

CD4(+) T cells that express the forkhead box P3 (FOXP3) transcription factor function as regulatory T (Treg) cells and hinder effective immune responses against cancer cells. Abundant Treg cell infiltration into tumors is associated with poor clinical outcomes in various types of cancers. However, the role of Treg cells is controversial in colorectal cancers (CRCs), in which FOXP3(+) T cell infiltration indicated better prognosis in some studies. Here we show that CRCs, which are commonly infiltrated by suppression-competent FOXP3(hi) Treg cells, can be classified into two types by the degree of additional infiltration of FOXP3(lo) nonsuppressive T cells. The latter, which are distinguished from FOXP3(+) Treg cells by non-expression of the naive T cell marker CD45RA and instability of FOXP3, secreted inflammatory cytokines. Indeed, CRCs with abundant infiltration of FOXP3(lo) T cells showed significantly better prognosis than those with predominantly FOXP3(hi) Treg cell infiltration. Development of such inflammatory FOXP3(lo) non-Treg cells may depend on secretion of interleukin (IL)-12 and transforming growth factor (TGF)-ß by tissues and their presence was correlated with tumor invasion by intestinal bacteria, especially Fusobacterium nucleatum. Thus, functionally distinct subpopulations of tumor-infiltrating FOXP3(+) T cells contribute in opposing ways to determining CRC prognosis. Depletion of FOXP3(hi) Treg cells from tumor tissues, which would augment antitumor immunity, could thus be used as an effective treatment strategy for CRCs and other cancers, whereas strategies that locally increase the population of FOXP3(lo) non-Treg cells could be used to suppress or prevent tumor formation.


Subject(s)
Colorectal Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Digestive System Surgical Procedures , Female , Forkhead Transcription Factors/metabolism , Humans , Interleukin-12 Subunit p35/immunology , Interleukin-12 Subunit p35/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
4.
Cell ; 163(2): 367-80, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26411289

ABSTRACT

Intestinal Th17 cells are induced and accumulate in response to colonization with a subgroup of intestinal microbes such as segmented filamentous bacteria (SFB) and certain extracellular pathogens. Here, we show that adhesion of microbes to intestinal epithelial cells (ECs) is a critical cue for Th17 induction. Upon monocolonization of germ-free mice or rats with SFB indigenous to mice (M-SFB) or rats (R-SFB), M-SFB and R-SFB showed host-specific adhesion to small intestinal ECs, accompanied by host-specific induction of Th17 cells. Citrobacter rodentium and Escherichia coli O157 triggered similar Th17 responses, whereas adhesion-defective mutants of these microbes failed to do so. Moreover, a mixture of 20 bacterial strains, which were selected and isolated from fecal samples of a patient with ulcerative colitis on the basis of their ability to cause a robust induction of Th17 cells in the mouse colon, also exhibited EC-adhesive characteristics.


Subject(s)
Bacterial Adhesion , Citrobacter rodentium/physiology , Enterobacteriaceae Infections/immunology , Escherichia coli Infections/immunology , Escherichia coli O157/physiology , Intestinal Mucosa/immunology , Th17 Cells/immunology , Animals , Bacterial Infections/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Feces/microbiology , Humans , Immunoglobulin A/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Microscopy, Electron, Scanning , Rats , Rats, Inbred F344 , Species Specificity
5.
Nature ; 500(7461): 232-6, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23842501

ABSTRACT

Manipulation of the gut microbiota holds great promise for the treatment of inflammatory and allergic diseases. Although numerous probiotic microorganisms have been identified, there remains a compelling need to discover organisms that elicit more robust therapeutic responses, are compatible with the host, and can affect a specific arm of the host immune system in a well-controlled, physiological manner. Here we use a rational approach to isolate CD4(+)FOXP3(+) regulatory T (Treg)-cell-inducing bacterial strains from the human indigenous microbiota. Starting with a healthy human faecal sample, a sequence of selection steps was applied to obtain mice colonized with human microbiota enriched in Treg-cell-inducing species. From these mice, we isolated and selected 17 strains of bacteria on the basis of their high potency in enhancing Treg cell abundance and inducing important anti-inflammatory molecules--including interleukin-10 (IL-) and inducible T-cell co-stimulator (ICOS)--in Treg cells upon inoculation into germ-free mice. Genome sequencing revealed that the 17 strains fall within clusters IV, XIVa and XVIII of Clostridia, which lack prominent toxins and virulence factors. The 17 strains act as a community to provide bacterial antigens and a TGF-ß-rich environment to help expansion and differentiation of Treg cells. Oral administration of the combination of 17 strains to adult mice attenuated disease in models of colitis and allergic diarrhoea. Use of the isolated strains may allow for tailored therapeutic manipulation of human immune disorders.


Subject(s)
Clostridium/immunology , Metagenome/immunology , T-Lymphocytes, Regulatory/physiology , Adult , Animals , Cell Proliferation , Clostridium/classification , Clostridium/genetics , Colitis/microbiology , Colitis/pathology , Colon/immunology , Colon/microbiology , Disease Models, Animal , Feces/microbiology , Germ-Free Life , Humans , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukin-10/metabolism , Male , Metagenome/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , RNA, Ribosomal, 16S/genetics , Rats , Rats, Inbred F344 , T-Lymphocytes, Regulatory/cytology
6.
Curr Opin Immunol ; 24(4): 392-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22673877

ABSTRACT

Foxp3+ CD4+ cells are prominent immune regulatory T (Treg) cells that are most abundant in the intestine. Recent studies have suggested that intestinal Treg cells consist of thymically and extrathymically developed cells that have unique characteristics. A fraction of intestinal Treg cells express T cell receptors that recognize antigens that are derived from the gut microbiota. The presence of the gut microbiota, particularly the Clostridium species, affects the development and function of Treg cells. These intestinal bacteria-induced Treg cells are likely to play a role in the tolerance toward the gut microbiota. These recent advances provide new insight into how T cells are educated in the intestine to maintain homeostasis with the gut microbiota.


Subject(s)
Clostridium/physiology , Intestinal Mucosa/microbiology , T-Lymphocytes, Regulatory/immunology , Animals , Colon/microbiology , Humans , Immune Tolerance , Metagenome
SELECTION OF CITATIONS
SEARCH DETAIL
...