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 ; 12(1): 3611, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34127673

ABSTRACT

Yeast is an integral part of mammalian microbiome, and like commensal bacteria, has the potential of being harnessed to influence immunity in clinical settings. However, functional specificities of yeast-derived immunoregulatory molecules remain elusive. Here we find that while under steady state, ß-1,3-glucan-containing polysaccharides potentiate pro-inflammatory properties, a relatively less abundant class of cell surface polysaccharides, dubbed mannan/ß-1,6-glucan-containing polysaccharides (MGCP), is capable of exerting potent anti-inflammatory effects to the immune system. MGCP, in contrast to previously identified microbial cell surface polysaccharides, through a Dectin1-Cox2 signaling axis in dendritic cells, facilitates regulatory T (Treg) cell induction from naïve T cells. Furthermore, through a TLR2-dependent mechanism, it restrains Th1 differentiation of effector T cells by suppressing IFN-γ expression. As a result, administration of MGCP display robust suppressive capacity towards experimental inflammatory disease models of colitis and experimental autoimmune encephalomyelitis (EAE) in mice, thereby highlighting its potential therapeutic utility against clinically relevant autoimmune diseases.


Subject(s)
Immunomodulation/drug effects , Immunomodulation/immunology , Polysaccharides/immunology , Saccharomyces cerevisiae/metabolism , beta-Glucans/immunology , Animals , CD4-Positive T-Lymphocytes , Cell Differentiation/drug effects , Colitis/immunology , Colitis/pathology , Cyclooxygenase 2 , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental , Glucans , Homeodomain Proteins/genetics , Immunity , Lectins, C-Type , Mannans , Mice , Mice, Inbred C57BL , Mice, Knockout , Polysaccharides/metabolism , Polysaccharides/pharmacology , Saccharomyces cerevisiae/genetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells , Zymosan , beta-Glucans/metabolism , beta-Glucans/pharmacology
2.
Sci Immunol ; 3(28)2018 10 19.
Article in English | MEDLINE | ID: mdl-30341145

ABSTRACT

Dysregulation of intestinal microflora is linked to inflammatory disorders associated with compromised immunosuppressive functions of Foxp3+ T regulatory (Treg) cells. Although mucosa-associated commensal microbiota has been implicated in Treg generation, molecular identities of the "effector" components controlling this process remain largely unknown. Here, we have defined Bifidobacterium bifidum as a potent inducer of Foxp3+ Treg cells with diverse T cell receptor specificity to dietary antigens, commensal bacteria, and B. bifidum itself. Cell surface ß-glucan/galactan (CSGG) polysaccharides of B. bifidum were identified as key components responsible for Treg induction. CSGG efficiently recapitulated the activity of whole bacteria and acted via regulatory dendritic cells through a partially Toll-like receptor 2-mediated mechanism. Treg cells induced by B. bifidum or purified CSGG display stable and robust suppressive capacity toward experimental colitis. By identifying CSGG as a functional component of Treg-inducing bacteria, our studies highlight the immunomodulatory potential of CSGG and CSGG-producing microbes.


Subject(s)
Bifidobacterium bifidum/immunology , Forkhead Transcription Factors/immunology , Polysaccharides/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bifidobacterium bifidum/cytology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Gastroenterology ; 152(8): 1998-2010, 2017 06.
Article in English | MEDLINE | ID: mdl-28246016

ABSTRACT

BACKGROUND & AIMS: Obesity and metabolic syndrome have been associated with alterations to the intestinal microbiota. However, few studies examined the effects of obesity on the intestinal immune system. We investigated changes in subsets of intestinal CD4+ T-helper (TH) cells with obesity and the effects of gut-tropic TH17 cells in mice on a high-fat diet (HFD). METHODS: We isolated immune cells from small intestine and adipose tissue of C57BL/6 mice fed a normal chow diet or a HFD for 10 weeks and analyzed the cells by flow cytometry. Mice fed a vitamin A-deficient HFD were compared with mice fed a vitamin A-sufficient HFD. Obese RAG1-deficient mice were given injections of only regulatory T cells or a combination of regulatory T cells and TH17 cells (wild type or deficient in integrin ß7 subunit or interleukin 17 [IL17]). Mice were examined for weight gain, fat mass, fatty liver, glucose tolerance, and insulin resistance. Fecal samples were collected before and after T cell transfer and analyzed for microbiota composition by metagenomic DNA sequencing and quantitative polymerase chain reaction. RESULTS: Mice placed on a HFD became obese, which affected the distribution of small intestinal CD4+ TH cells. Intestinal tissues from obese mice had significant reductions in the proportion of TH17 cells but increased proportion of TH1 cells, compared with intestinal tissues from nonobese mice. Depletion of vitamin A in obese mice further reduced the proportion of TH17 cells in small intestine; this reduction correlated with more weight gain and worsening of glucose intolerance and insulin resistance. Adoptive transfer of in vitro-differentiated gut-tropic TH17 cells to obese mice reduced these metabolic defects, which required the integrin ß7 subunit and IL17. Delivery of TH17 cells to intestines of mice led to expansion of commensal microbes associated with leanness. CONCLUSIONS: In mice, intestinal TH17 cells contribute to development of a microbiota that maintains metabolic homeostasis, via IL17. Gut-homing TH17 cells might be used to reduce metabolic disorders in obese individuals.


Subject(s)
Adoptive Transfer , Immunity, Mucosal , Insulin Resistance , Intestine, Small/immunology , Metabolic Syndrome/prevention & control , Obesity/prevention & control , Th17 Cells/transplantation , Animals , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Feces/microbiology , Gastrointestinal Microbiome/immunology , Genotype , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Host-Pathogen Interactions , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Interleukin-17/deficiency , Interleukin-17/genetics , Intestine, Small/metabolism , Intestine, Small/microbiology , Male , Metabolic Syndrome/genetics , Metabolic Syndrome/immunology , Metabolic Syndrome/microbiology , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/immunology , Obesity/microbiology , Phenotype , Th17 Cells/immunology , Th17 Cells/microbiology , Time Factors , Vitamin A Deficiency/complications
4.
J Exp Med ; 213(4): 555-67, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-26951334

ABSTRACT

Eosinophils play proinflammatory roles in helminth infections and allergic diseases. Under steady-state conditions, eosinophils are abundantly found in the small intestinal lamina propria, but their physiological function is largely unexplored. In this study, we found that small intestinal eosinophils down-regulate Th17 cells. Th17 cells in the small intestine were markedly increased in the ΔdblGATA-1 mice lacking eosinophils, and an inverse correlation was observed between the number of eosinophils and that of Th17 cells in the small intestine of wild-type mice. In addition, small intestinal eosinophils suppressed the in vitro differentiation of Th17 cells, as well as IL-17 production by small intestinal CD4(+)T cells. Unlike other small intestinal immune cells or circulating eosinophils, we found that small intestinal eosinophils have a unique ability to constitutively secrete high levels of IL-1 receptor antagonist (IL-1Ra), a natural inhibitor of IL-1ß. Moreover, small intestinal eosinophils isolated from IL-1Ra-deficient mice failed to suppress Th17 cells. Collectively, our results demonstrate that small intestinal eosinophils play a pivotal role in the maintenance of intestinal homeostasis by regulating Th17 cells via production of IL-1Ra.


Subject(s)
Cystinyl Aminopeptidase/immunology , Eosinophils/immunology , Intestine, Small/immunology , Th17 Cells/immunology , Animals , Cystinyl Aminopeptidase/genetics , Eosinophils/cytology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Intestine, Small/cytology , Mice , Mice, Transgenic , Th17 Cells/cytology
5.
J Allergy Clin Immunol ; 137(2): 507-516.e8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26433560

ABSTRACT

BACKGROUND: The incidence of food allergies has increased dramatically during the last decade. Recently, probiotics have been studied for the prevention and treatment of allergic disease. OBJECTIVE: We examined whether Bifidobacterium longum KACC 91563 and Enterococcus faecalis KACC 91532 have the capacity to suppress food allergies. METHODS: B longum KACC 91563 and E faecalis KACC 91532 were administered to BALB/c wild-type mice, in which food allergy was induced by using ovalbumin and alum. Food allergy symptoms and various immune responses were assessed. RESULTS: B longum KACC 91563, but not E faecalis KACC 91532, alleviated food allergy symptoms. Extracellular vesicles of B longum KACC 91563 bound specifically to mast cells and induced apoptosis without affecting T-cell immune responses. Furthermore, injection of family 5 extracellular solute-binding protein, a main component of extracellular vesicles, into mice markedly reduced the occurrence of diarrhea in a mouse food allergy model. CONCLUSION: B longum KACC 91563 induces apoptosis of mast cells specifically and alleviates food allergy symptoms. Accordingly, B longum KACC 91563 and family 5 extracellular solute-binding protein exhibit potential as therapeutic approaches for food allergies.


Subject(s)
Bacterial Proteins/immunology , Bifidobacterium/immunology , Extracellular Vesicles/immunology , Food Hypersensitivity/immunology , Food Hypersensitivity/microbiology , Immunomodulation , Mast Cells/immunology , Animals , Apoptosis/immunology , Bacterial Proteins/metabolism , Bifidobacterium/metabolism , Cell Count , Cytokines/biosynthesis , Disease Models, Animal , Endocytosis/immunology , Extracellular Vesicles/metabolism , Food Hypersensitivity/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/microbiology , Intestine, Small/pathology , Mast Cells/metabolism , Mast Cells/microbiology , Mice , Probiotics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
6.
PLoS One ; 8(10): e76716, 2013.
Article in English | MEDLINE | ID: mdl-24116141

ABSTRACT

The bark of Ulmus davidiana var. japonica Nakai (Ulmaceae) has been used in traditional Korean medicine for chronic inflammation in the gastrointestinal tract. Here we investigated the frequency and cytokine profile of the major immune cells in the small intestinal lamina propria (SI LP), spleen, and mesenteric lymph nodes (MLNs) of mice treated orally with Ulmus davidiana var. japonica Nakai bark water extract (UDE) to address the immunomodulatory role of this herb in intestinal homeostasis. B6 mice were given 5g/kg UDE once daily for 14 days. They were then sacrificed, and cells were isolated from the spleen, MLNs, and SI LP. The proportion of B versus T lymphocytes, CD4(+) versus CD8(+) T lymphocytes, Th1 and Th17 cells, and Foxp3(+) regulatory T cells in the spleen, MLNs, and SI LP were analyzed. The frequency of antigen-presenting cells (APCs), including dendritic cells, macrophages, and eosinophils in the SI LP and the expression of costimulatory molecules on APCs were also evaluated. The numbers and frequencies of Th1 and Th17 cells in the SI LP were significantly reduced in the UDE-treated mice compared with PBS controls. In addition, the proportion of IL-4-producing eosinophils in the SI LP was significantly elevated in the UDE-treated mice compared with controls. Taken together, these data indicate that UDE up-regulates the number and frequency of SI LP eosinophils, which can down-regulate the Th1 and Th17 responses via IL-4 secretion and contribute to intestinal homeostasis.


Subject(s)
Eosinophils/drug effects , Intestine, Small/drug effects , Plant Extracts/pharmacology , Th1 Cells/drug effects , Th17 Cells/drug effects , Ulmus/chemistry , Administration, Oral , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Count , Enzyme-Linked Immunosorbent Assay , Eosinophils/immunology , Eosinophils/metabolism , Female , Homeostasis/drug effects , Homeostasis/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interleukin-4/immunology , Interleukin-4/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plant Bark/chemistry , Plant Extracts/administration & dosage , Plant Extracts/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...