Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Inflammation ; 45(1): 372-386, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34792688

RESUMO

Intestinal inflammation is a common disease which can further lead to inflammatory bowel disease and even intestinal cancer. The increasing focus has come to the role of short-chain fatty acid (SCFA) in various bowel diseases. Hence, this study was designed to explore the specific role of SCFA in intestinal inflammation. In vivo and in vitro models of intestinal inflammation were constructed by lipopolysaccharide (LPS) injection in mice and LPS treatment on intestinal epithelial cells. A possible regulatory mechanism involving SCFA, CCAAT enhancer-binding protein beta (CEBPB), microRNA-145 (miR-145), and dual-specificity phosphatase 6 (DUSP6) in intestinal inflammation was verified by ChIP assay and dual-luciferase reporter gene assay. To evaluate the effects of SCFA on LPS-treated intestinal epithelial cells, the expression of relevant genes and inflammatory factors (IL-6, TNF-α, and IL-1ß) were determined. Last, the role of SCFA in vivo was explored through the scoring of disease activity index (DAI) and observation of colonic histology of LPS-treated mice. SCFA decreased the CEBPB expression in mouse colon tissues and small intestine epithelial cells induced by LPS. Furthermore, CEBPB could bind to the miR-145 promoter to inhibit its expression, thereby promoting the expression of DUSP6. In addition, SCFA improved the DAI, colonic histology, and the expression of serum inflammatory factors in LPS-treated mice and cells, noting that SCFA alleviated intestinal inflammation in vitro and in vivo. To sum up, SCFA inhibited DUSP6 by upregulating miR-145 through CEBPB repression and thus prevented the development of intestinal inflammation.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Colite/metabolismo , Colo/metabolismo , Fosfatase 6 de Especificidade Dupla/metabolismo , Ácidos Graxos Voláteis/metabolismo , Mucosa Intestinal/metabolismo , MicroRNAs/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/imunologia , Colite/imunologia , Colite/patologia , Colo/imunologia , Colo/patologia , Fosfatase 6 de Especificidade Dupla/imunologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Ácidos Graxos Voláteis/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/imunologia
2.
Int J Mol Sci ; 22(16)2021 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-34445184

RESUMO

The microbiome, i.e., the communities of microbes that inhabit the surfaces exposed to the external environment, participates in the regulation of host physiology, including the immune response against pathogens. At the same time, the immune response shapes the microbiome to regulate its composition and function. How the crosstalk between the immune system and the microbiome regulates the response to fungal infection has remained relatively unexplored. We have previously shown that strict anaerobes protect from infection with the opportunistic fungus Aspergillus fumigatus by counteracting the expansion of pathogenic Proteobacteria. By resorting to immunodeficient mouse strains, we found that the lung microbiota could compensate for the lack of B and T lymphocytes in Rag1-/- mice by skewing the composition towards an increased abundance of protective anaerobes such as Clostridia and Bacteroidota. Conversely, NSG mice, with major defects in both the innate and adaptive immune response, showed an increased susceptibility to infection associated with a low abundance of strict anaerobes and the expansion of Proteobacteria. Further exploration in a murine model of chronic granulomatous disease, a primary form of immunodeficiency characterized by defective phagocyte NADPH oxidase, confirms the association of lung unbalance between anaerobes and Proteobacteria and the susceptibility to aspergillosis. Consistent changes in the lung levels of short-chain fatty acids between the different strains support the conclusion that the immune system and the microbiota are functionally intertwined during Aspergillus infection and determine the outcome of the infection.


Assuntos
Aspergilose/imunologia , Aspergillus fumigatus/imunologia , Pulmão/microbiologia , Imunidade Adaptativa , Animais , Aspergilose/microbiologia , Aspergillus fumigatus/fisiologia , Ácidos Graxos Voláteis/imunologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microbiota
3.
Biomed Pharmacother ; 140: 111542, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34088571

RESUMO

Recent studies report that the gut microbiome can enhance systemic and antitumor immunity by modulating responses to antibody immunotherapy in melanoma patients. In this study, we found that icariside I, a novel anti-cancer agent isolated from Epimedium, significantly inhibited B16F10 melanoma growth in vivo through regulation of gut microbiota and host immunity. Oral administration of icariside I improved the microbiota community structure with marked restoration of Lactobacillus spp. and Bifidobacterium spp. abundance in the cecal contents of tumor-bearing mice. We also found that icariside I improves the levels of microbiota-derived metabolites such as short-chain fatty acids (SCFAs) and indole derivatives, consequently promoting repair of the intestinal barrier and reducing systemic inflammation of tumor-bearing mice. Icariside I exhibited strong immunological anti-tumor activity, directly manifested by up-regulation of multiple lymphocyte subsets including CD4+ and CD8+ T cells or NK and NKT cells in peripheral blood of tumor-bearing mice. Collectively, these results suggest that icariside I, via its microbiome remodeling and host immune regulation properties, may be developed as an anticancer drug.


Assuntos
Antineoplásicos/farmacologia , Flavonas/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Melanoma/imunologia , Melanoma/terapia , Microbiota/efeitos dos fármacos , Umbeliferonas/farmacologia , Animais , Ceco/microbiologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ácidos Graxos Voláteis/imunologia , Fezes/microbiologia , Feminino , Imunoterapia/métodos , Indóis/farmacologia , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
4.
Infect Immun ; 89(9): e0018821, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34097474

RESUMO

Short-chain fatty acids (SCFAs) are the main metabolites produced by the gut microbiota via the fermentation of complex carbohydrates and fibers. Evidence suggests that SCFAs play a role in the control of infections through direct action both on microorganisms and on host signaling. This review summarizes the main microbicidal effects of SCFAs and discusses studies highlighting the effect of SCFAs in the virulence and viability of microorganisms. We also describe the diverse and complex modes of action of the SCFAs on the immune system in the face of infections with a specific focus on bacterial and viral respiratory infections. A growing body of evidence suggests that SCFAs protect against lung infections. Finally, we present potential strategies that may be leveraged to exploit the biological properties of SCFAs for increasing effectiveness and optimizing patient benefits.


Assuntos
Anti-Infecciosos/uso terapêutico , Ácidos Graxos Voláteis/uso terapêutico , Infecções/tratamento farmacológico , Pulmão/efeitos dos fármacos , Animais , Anti-Infecciosos/imunologia , Anti-Infecciosos/metabolismo , Ácidos Graxos Voláteis/imunologia , Ácidos Graxos Voláteis/metabolismo , Humanos , Infecções/imunologia , Infecções/microbiologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/virologia , Viabilidade Microbiana , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/imunologia , Infecções Respiratórias/microbiologia , Infecções Respiratórias/virologia , Transdução de Sinais/imunologia , Virulência
5.
Biomed Pharmacother ; 141: 111817, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34126349

RESUMO

In recent years, an overwhelming amount of evidence has positively recommended a significant role of microbiota in human health and disease. Microbiota also plays a crucial role in the initiation, preparation, and function of the host immune response. Recently, it has been shown that short-chain fatty acids (SCFAs) are the primary metabolites of the intestinal microbiota produced by anaerobic fermentation, which contributes to the host-pathogen interaction. SCFAs, such as propionate, acetate, and butyrate, are bacterial metabolites with immunomodulatory activity, and they are indispensable for the maintenance of homeostasis. Some evidence indicates that they are involved in the development of infections. In the present study, we provide the latest findings on the role of SCFAs in response to bacterial infections.


Assuntos
Infecções Bacterianas/imunologia , Ácidos Graxos Voláteis/farmacologia , Fatores Imunológicos/farmacologia , Microbiota , Animais , Ácidos Graxos Voláteis/imunologia , Microbioma Gastrointestinal , Humanos
6.
Gut Microbes ; 13(1): 1-22, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33590776

RESUMO

The interaction disorder between gut microbiota and its host has been documented in different non-communicable diseases (NCDs) such as metabolic syndrome, neurodegenerative disease, and autoimmune disease. The majority of these altered interactions arise through metabolic cross-talk between gut microbiota and host immune system, inducing a low-grade chronic inflammation that characterizes all NCDs. In this review, we discuss the contribution of bacterial metabolites to immune signaling pathways involved in NCDs. We then review recent advances that aid to rationally design microbial therapeutics. A deeper understanding of these intersections between host and gut microbiota metabolism using metabolomics-based system biology platform promises to reveal the fundamental mechanisms that drive metabolic predispositions to disease and suggest new avenues to use microbial therapeutic opportunities for NCDs treatment and prevention. Abbreviations: NCDs: non-communicable disease, IBD: inflammatory bowel disease, IL: interleukin, T2D: type 2 diabetes, SCFAs: short-chain fatty acids, HDAC: histone deacetylases, GPCR: G-protein coupled receptors, 5-HT: 5-hydroxytryptamine receptor signaling, DCs: dendritic cells, IECs: intestinal epithelial cells, T-reg: T regulatory cell, NF-κB: nuclear factor κB, TNF-α: tumor necrosis factor alpha, Th: T helper cell, CNS: central nervous system, ECs: enterochromaffin cells, NSAIDs: non-steroidal anti-inflammatory drugs, AhR: aryl hydrocarbon receptor, IDO: indoleamine 2,3-dioxygenase, QUIN: quinolinic acid, PC: phosphatidylcholine, TMA: trimethylamine, TMAO: trimethylamine N-oxide, CVD: cardiovascular disease, NASH: nonalcoholic steatohepatitis, BAs: bile acids, FXR: farnesoid X receptor, CDCA: chenodeoxycholic acid, DCA: deoxycholic acid, LCA: lithocholic acid, UDCA: ursodeoxycholic acid, CB: cannabinoid receptor, COBRA: constraint-based reconstruction and analysis.


Assuntos
Bactérias/metabolismo , Microbioma Gastrointestinal/fisiologia , Doenças não Transmissíveis , Transdução de Sinais/imunologia , Amidas/imunologia , Amidas/metabolismo , Bactérias/classificação , Bactérias/isolamento & purificação , Ácidos e Sais Biliares/imunologia , Ácidos e Sais Biliares/metabolismo , Colina/imunologia , Colina/metabolismo , Suscetibilidade a Doenças/imunologia , Suscetibilidade a Doenças/microbiologia , Ácidos Graxos Voláteis/imunologia , Ácidos Graxos Voláteis/metabolismo , Humanos , Sistema Imunitário/imunologia , Indóis/imunologia , Indóis/metabolismo , Poliaminas/imunologia , Poliaminas/metabolismo , Vitaminas/imunologia , Vitaminas/metabolismo
7.
Cells ; 10(1)2021 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-33435197

RESUMO

Although the etiology of multiple sclerosis (MS) is still unknown, it is commonly accepted that environmental factors could contribute to the disease. The objective of this study was to analyze the humoral response to Epstein-Barr virus, human herpesvirus 6A/B and cytomegalovirus, and the levels of 25-hydroxyvitamin D (25(OH)D) and the three main short-chain fatty acids (SCFA), propionate (PA), butyrate (BA) and acetate (AA), in MS patients and healthy controls (HC) to understand how they could contribute to the pathogenesis of the disease. With this purpose, we analyzed the correlations among them and with different clinical variables and a wide panel of cell subsets. We found statistically significant differences for most of the environmental factors analyzed when we compared MS patients and HC, supporting their possible involvement in the disease. The strongest correlations with the clinical variables and the cell subsets analyzed were found for 25(OH)D and SCFAs levels. A correlation was also found between 25(OH)D and PA/AA ratio, and the interaction between these factors negatively correlated with interleukin 17 (IL-17)-producing CD4+ and CD8+ T cells in untreated MS patients. Therapies that simultaneously increase vitamin D levels and modify the proportion of SCFA could be evaluated in the future.


Assuntos
Anticorpos Antivirais/imunologia , Ácidos Graxos Voláteis/imunologia , Herpesvirus Humano 4/imunologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Vitamina D/metabolismo , Adulto , Estudos de Casos e Controles , Meio Ambiente , Feminino , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Masculino , Pessoa de Meia-Idade , Vitamina D/análogos & derivados
8.
J Infect Dis ; 223(12 Suppl 2): S194-S200, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33326565
9.
Front Immunol ; 12: 761820, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35069532

RESUMO

Dietary supplementation of fish with ß-glucans has been commonly associated with immunomodulation and generally accepted as beneficial for fish health. However, to date the exact mechanisms of immunomodulation by ß-glucan supplementation in fish have remained elusive. In mammals, a clear relation between high-fibre diets, such as those including ß-glucans, and diet-induced immunomodulation via intestinal microbiota and associated metabolites has been observed. In this study, first we describe by 16S rRNA sequencing the active naive microbiota of common carp intestine. Based on the abundance of the genus Bacteroides, well known for their capacity to degrade and ferment carbohydrates, we hypothesize that common carp intestinal microbiota could ferment dietary ß-glucans. Indeed, two different ß-glucan preparations (curdlan and MacroGard®) were both fermented in vitro, albeit with distinct fermentation dynamics and distinct production of short-chain fatty acids (SCFA). Second, we describe the potential immunomodulatory effects of the three dominant SCFAs (acetate, butyrate, and propionate) on head kidney leukocytes, showing effects on both nitric oxide production and expression of several cytokines (il-1b, il-6, tnfα, and il-10) in vitro. Interestingly, we also observed a regulation of expression of several gpr40L genes, which were recently described as putative SCFA receptors. Third, we describe how a single in vivo oral gavage of carp with MacroGard® modulated simultaneously, the expression of several pro-inflammatory genes (il-1b, il-6, tnfα), type I IFN-associated genes (tlr3.1, mx3), and three specific gpr40L genes. The in vivo observations provide indirect support to our in vitro data and the possible role of SCFAs in ß-glucan-induced immunomodulation. We discuss how ß-glucan-induced immunomodulatory effects can be explained, at least in part, by fermentation of MacroGard® by specific bacteria, part of the naive microbiota of common carp intestine, and how a subsequent production of SFCAs could possibly explain immunomodulation by ß-glucan via SCFA receptors present on leukocytes.


Assuntos
Ração Animal , Carpas , Ácidos Graxos Voláteis/imunologia , Microbioma Gastrointestinal , Imunomodulação/efeitos dos fármacos , beta-Glucanas/farmacologia , Animais , Carpas/imunologia , Carpas/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia
10.
Acta Biochim Pol ; 67(4): 485-493, 2020 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-33332076

RESUMO

Exopolysaccharides (EPSs) possess many bioactivities such as immune regulation, antioxidant, anti-tumor and modulation of intestinal microbial balance but their direct effect on inflammatory bowel disease (IBD) response has not been studied. The purpose of this study was to evaluate the anti-inflammatory effect of EPS produced by L. plantarum YW11 administered at different dosages in IBD mouse model induced with 5% dextran sulphate sodium (DSS). The DSS-induced colitis, accompanied by body weight loss, reduction of colon coefficient and histological colon injury was considerably ameliorated in mice fed the EPS (10 mg/kg). The middle dose of the EPS (25 mg/kg) could effectively recover the intestinal microbial diversity and increase the abundance of Roseburia, Ruminococcus and Blautia with increased content of butyric acid. Moreover, EPS also reduced the production of pro-inflammatory cytokines (TNF-α, IL-1ß, IL-6, IFN-γ, IL-12 and IL-18) and enhanced the anti-inflammatory cytokine IL-10. This study showed that EPS might help in modulation of gut microbiota and improve the immunity of the host to reduce the risk of IBD symptoms.


Assuntos
Anti-Inflamatórios/farmacologia , Colite/tratamento farmacológico , Colo/imunologia , Microbioma Gastrointestinal/imunologia , Lactobacillus plantarum/química , Polissacarídeos Bacterianos/farmacologia , Animais , Anti-Inflamatórios/isolamento & purificação , Peso Corporal/efeitos dos fármacos , Clostridiales , Colite/induzido quimicamente , Colite/imunologia , Colite/microbiologia , Colo/metabolismo , Colo/microbiologia , Sulfato de Dextrana , Ácidos Graxos Voláteis/imunologia , Ácidos Graxos Voláteis/metabolismo , Fezes/microbiologia , Expressão Gênica , Imunidade Inata , Interferon gama/genética , Interferon gama/imunologia , Interleucinas/genética , Interleucinas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Polissacarídeos Bacterianos/isolamento & purificação , Prednisolona/análogos & derivados , Prednisolona/farmacologia , RNA Ribossômico 16S/genética , Ruminococcus , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
11.
Front Immunol ; 11: 590685, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33363537

RESUMO

Converging evidences showed that people with diabetes mellitus (DM) have significantly higher risk for different cancers, of which the exact mechanism underlying the association has not been fully realized. Short-chain fatty acids (SCFAs), the fermentation products of the intestinal microbiota, are an essential source for energy supply in gut epithelial cells. They have been reported to improve intestinal barrier integrity, prevent microbial translocation, and further dampen inflammation. Gut dysbiosis and reduction in SCFA-producing bacteria as well as SCFAs production in the intestine are commonly seen in metabolic disorders including DM and obesity. Moreover, inflammation can contribute to tumor initiation and progression through multiple pathways, such as enhancing DNA damage, accumulating mutations in tumor suppressor genes Tp53, and activating nuclear factor-kappa B (NF-κB) signaling pathways. Based on these facts, we hypothesize that lower levels of microbial SCFAs resulted from gut dysbiosis in diabetic individuals, enhance microbial translocation, and increase the inflammatory responses, inducing tumorigenesis ulteriorly. To this end, we will discuss protective properties of microbial SCFAs and explore the pivotal roles SCFAs played in the link of DM with cancer, so as to take early precautions to reduce the risk of cancer in patients with DM.


Assuntos
Carcinogênese/imunologia , Diabetes Mellitus/imunologia , Ácidos Graxos Voláteis/imunologia , Inflamação/imunologia , Animais , Diabetes Mellitus/microbiologia , Disbiose/imunologia , Disbiose/microbiologia , Microbioma Gastrointestinal , Humanos , Inflamação/microbiologia , Neoplasias/imunologia , Neoplasias/microbiologia
12.
Int J Mol Sci ; 21(22)2020 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-33227973

RESUMO

Inflammation is the key for the initiation and progression of atherosclerosis. Accumulating evidence has revealed that an altered gut microbiome (dysbiosis) triggers both local and systemic inflammation to cause chronic inflammatory diseases, including atherosclerosis. There have been some microbiome-relevant pro-inflammatory mechanisms proposed to link the relationships between dysbiosis and atherosclerosis such as gut permeability disruption, trigger of innate immunity from lipopolysaccharide (LPS), and generation of proatherogenic metabolites, such as trimethylamine N-oxide (TMAO). Meanwhile, immune responses, such as inflammasome activation and cytokine production, could reshape both composition and function of the microbiota. In fact, the immune system delicately modulates the interplay between microbiota and atherogenesis. Recent clinical trials have suggested the potential of immunomodulation as a treatment strategy of atherosclerosis. Here in this review, we present current knowledge regarding to the roles of microbiota in contributing atherosclerotic pathogenesis and highlight translational perspectives by discussing the mutual interplay between microbiota and immune system on atherogenesis.


Assuntos
Aterosclerose/imunologia , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Imunidade Inata , Imunomodulação , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/microbiologia , Aterosclerose/patologia , Ensaios Clínicos como Assunto , Citocinas/imunologia , Citocinas/metabolismo , Progressão da Doença , Disbiose/tratamento farmacológico , Disbiose/microbiologia , Disbiose/patologia , Ácidos Graxos Voláteis/imunologia , Ácidos Graxos Voláteis/metabolismo , Humanos , Fatores Imunológicos/uso terapêutico , Inflamassomos/imunologia , Inflamassomos/metabolismo , Inflamação , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Metilaminas/imunologia , Metilaminas/metabolismo
13.
Nat Commun ; 11(1): 4457, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32901017

RESUMO

Innate lymphoid cells (ILCs) and CD4+ T cells produce IL-22, which is critical for intestinal immunity. The microbiota is central to IL-22 production in the intestines; however, the factors that regulate IL-22 production by CD4+ T cells and ILCs are not clear. Here, we show that microbiota-derived short-chain fatty acids (SCFAs) promote IL-22 production by CD4+ T cells and ILCs through G-protein receptor 41 (GPR41) and inhibiting histone deacetylase (HDAC). SCFAs upregulate IL-22 production by promoting aryl hydrocarbon receptor (AhR) and hypoxia-inducible factor 1α (HIF1α) expression, which are differentially regulated by mTOR and Stat3. HIF1α binds directly to the Il22 promoter, and SCFAs increase HIF1α binding to the Il22 promoter through histone modification. SCFA supplementation enhances IL-22 production, which protects intestines from inflammation. SCFAs promote human CD4+ T cell IL-22 production. These findings establish the roles of SCFAs in inducing IL-22 production in CD4+ T cells and ILCs to maintain intestinal homeostasis.


Assuntos
Ácidos Graxos Voláteis/imunologia , Microbioma Gastrointestinal/imunologia , Imunidade Inata , Interleucinas/biossíntese , Animais , Butiratos/imunologia , Butiratos/metabolismo , Butiratos/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Citrobacter rodentium , Colite/imunologia , Colite/microbiologia , Colite/prevenção & controle , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/prevenção & controle , Ácidos Graxos Voláteis/metabolismo , Ácidos Graxos Voláteis/farmacologia , Microbioma Gastrointestinal/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Técnicas In Vitro , Interleucinas/deficiência , Interleucinas/genética , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Regiões Promotoras Genéticas , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Interleucina 22
14.
Front Immunol ; 11: 1427, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32903327

RESUMO

Short chain fatty acids (SFCAs) are microbial metabolites produced in the gut upon fermentation of dietary fiber. These metabolites interact with the host immune system and can elicit epigenetic effects. There is evidence to suggest that SCFAs may play a role in the developmental programming of immune disorders and obesity, though evidence in humans remains sparse. Here we have quantified human milk (HM) SCFA levels in an international cohort of atopic and non-atopic mothers (n = 109). Our results demonstrate that human milk contains detectable levels of the SCFAs acetate, butyrate, and formate. Samples from atopic mothers had significantly lower concentrations of acetate and butyrate than those of non-atopic mothers. HM SCFA levels in atopic and non-atopic women also varied based on maternal country of residence (Australia, Japan, Norway, South Africa, USA). Reduced exposure to HM SCFA in early life may program atopy or overweight risk in breastfed infants.


Assuntos
Ácidos Graxos Voláteis , Hipersensibilidade , Leite Humano/química , Adulto , Ácidos Graxos Voláteis/imunologia , Feminino , Humanos , Hipersensibilidade/imunologia , Lactente , Leite Humano/metabolismo , Mães
15.
Int J Mol Sci ; 21(14)2020 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-32664466

RESUMO

Short-chain fatty acids (SCFA), bacterial metabolites released from dental biofilm, are supposed to target the oral epithelium. There is, however, no consensus on how SCFA affect the oral epithelial cells. The objective of the present study was to systematically review the available in vitro evidence of the impact of SCFA on human oral epithelial cells in the context of periodontal disease. A comprehensive electronic search using five databases along with a grey literature search was performed. In vitro studies that evaluated the effects of SCFA on human oral epithelial cells were eligible for inclusion. Risk of bias was assessed by the University of Bristol's tool for assessing risk of bias in cell culture studies. Certainty in cumulative evidence was evaluated using GRADE criteria (grading of recommendations assessment, development, and evaluation). Of 3591 records identified, 10 were eligible for inclusion. A meta-analysis was not possible due to the heterogeneity between the studies. The risk of bias across the studies was considered "serious" due to the presence of methodological biases. Despite these limitations, this review showed that SCFA negatively affect the viability of oral epithelial cells by activating a series of cellular events that includes apoptosis, autophagy, and pyroptosis. SCFA impair the integrity and presumably the transmigration of leucocytes through the epithelial layer by changing junctional and adhesion protein expression, respectively. SCFA also affect the expression of chemokines and cytokines in oral epithelial cells. Future research needs to identify the underlying signaling cascades and to translate the in vitro findings into preclinical models.


Assuntos
Disbiose/complicações , Células Epiteliais/efeitos dos fármacos , Ácidos Graxos Voláteis/efeitos adversos , Mucosa Bucal/microbiologia , Periodontite/etiologia , Apoptose/efeitos dos fármacos , Biofilmes , Butiratos/farmacologia , Adesão Celular/efeitos dos fármacos , Forma Celular , Disbiose/microbiologia , Ácidos Graxos Voláteis/imunologia , Humanos , Microbiota , Mucosa Bucal/citologia , Bolsa Periodontal/microbiologia , Periodontite/tratamento farmacológico , Bicarbonato de Sódio/farmacologia , Bicarbonato de Sódio/uso terapêutico
16.
J Am Soc Nephrol ; 31(7): 1445-1461, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32482686

RESUMO

BACKGROUND: Short-chain fatty acids derived from gut microbial fermentation of dietary fiber have been shown to suppress autoimmunity through mechanisms that include enhanced regulation by T regulatory cells (Tregs). METHODS: Using a murine kidney transplantation model, we examined the effects on alloimmunity of a high-fiber diet or supplementation with the short-chain fatty acid acetate. Kidney transplants were performed from BALB/c(H2d) to B6(H2b) mice as allografts in wild-type and recipient mice lacking the G protein-coupled receptor GPR43 (the metabolite-sensing receptor of acetate). Allograft mice received normal chow, a high-fiber diet, or normal chow supplemented with sodium acetate. We assessed rejection at days 14 (acute) and 100 (chronic), and used 16S rRNA sequencing to determine gut microbiota composition pretransplantation and post-transplantation. RESULTS: Wild-type mice fed normal chow exhibited dysbiosis after receiving a kidney allograft but not an isograft, despite the avoidance of antibiotics and immunosuppression for the latter. A high-fiber diet prevented dysbiosis in allograft recipients, who demonstrated prolonged survival and reduced evidence of rejection compared with mice fed normal chow. Allograft mice receiving supplemental sodium acetate exhibited similar protection from rejection, and subsequently demonstrated donor-specific tolerance. Depletion of CD25+ Tregs or absence of the short-chain fatty acid receptor GPR43 abolished this survival advantage. CONCLUSIONS: Manipulation of the microbiome by a high-fiber diet or supplementation with sodium acetate modified alloimmunity in a kidney transplant model, generating tolerance dependent on Tregs and GPR43. Diet-based therapy to induce changes in the gut microbiome can alter systemic alloimmunity in mice, in part through the production of short-chain fatty acids leading to Treg cell development, and merits study as a potential clinical strategy to facilitate transplant acceptance.


Assuntos
Fibras na Dieta/administração & dosagem , Ácidos Graxos Voláteis/imunologia , Microbioma Gastrointestinal/imunologia , Rejeição de Enxerto/prevenção & controle , Tolerância Imunológica/efeitos dos fármacos , Linfócitos T Reguladores , Doença Aguda , Aloenxertos/imunologia , Animais , Ácido Butírico/farmacologia , Doença Crônica , Suplementos Nutricionais , Disbiose/etiologia , Disbiose/microbiologia , Disbiose/prevenção & controle , Microbioma Gastrointestinal/efeitos dos fármacos , Rejeição de Enxerto/patologia , Rejeição de Enxerto/fisiopatologia , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/imunologia , Transplante de Rim/efeitos adversos , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Acetato de Sódio/farmacologia
17.
Artigo em Inglês | MEDLINE | ID: mdl-32413493

RESUMO

This study was to explore the impacts of water-soluble chitosan and mixed probiotics on growth performance, intestinal short-chain fatty acids (SCFAs) and immunity and ammonia resistance in Litopenaeus vannamei. Shrimp were fed one of four experimental diets including basal diet (CON), 0.10% water-soluble chitosan diet (WSC), 0.30% mixed probiotics (MP) and 0.10% water-soluble chitosan +0.30% mixed probiotics (SYN) for 8 weeks. Results showed shrimp fed with dietary MP and SYN diets could significantly improve growth performance and feed utilization in comparison with those of shrimp fed with CON diet (P < 0.05). Acetic acid content was significantly higher in shrimp fed with all supplemented diets compared to that in shrimp fed with CON diet (P < 0.05). Compared to shrimp fed with CON diet, dietary WSC and MP significantly influenced the contents and/or activities of aspartate aminotransferase (AST), total protein (TP), superoxide dismutase (SOD), lysozyme (LZM) in serum, SOD, malondialdehyde (MDA), acid phosphatase (ACP) in hepatopancreas and SOD and MDA in intestine. In addition, the gene expression levels of prophenoloxidase (proPO), penaiedin 3a (Pen-3a), crustin (Crustin), serine proteinase (SP), GPX and SOD in hepatopancreas, were significantly upregulated compared to those in CON diet at some time points (P < 0.05). Significantly higher survival rate in all supplemented diets were observed after ammonia challenge (P < 0.05). Therefore, the above results indicated dietary WSC and MP or SYN could enhance intestinal SCFAs content, stimulated antioxidant capacity and immune response, and increase the ammonia resistance of Litopenaeus vannamei. Besides, the growth performance was also improved by dietary MP and SYN.


Assuntos
Quitosana/administração & dosagem , Ácidos Graxos Voláteis/imunologia , Penaeidae/imunologia , Probióticos/administração & dosagem , Amônia/toxicidade , Ração Animal , Animais , Suplementos Nutricionais , Ácidos Graxos Voláteis/metabolismo , Imunidade Inata/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/imunologia , Penaeidae/efeitos dos fármacos , Penaeidae/crescimento & desenvolvimento , Penaeidae/metabolismo
18.
JCI Insight ; 5(10)2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32298241

RESUMO

The incidence of type 1 diabetes (T1D) has been increasing among children and adolescents, in which environmental factors, including gut microbiota, play an important role. However, the underlying mechanisms are yet to be determined. Here, we show that patients with newly diagnosed T1D displayed not only a distinct profile of gut microbiota associated with decreased short-chain fatty acids (SCFAs) production, but also an altered IgA-mediated immunity compared with healthy control subjects. Using germ-free NOD mice, we demonstrate that gut microbiota from patients with T1D promoted different IgA-mediated immune responses compared with healthy control gut microbiota. Treatment with the SCFA, acetate, reduced gut bacteria-induced IgA response accompanied by decreased severity of insulitis in NOD mice. We believe our study provides new insights into the functional effects of gut microbiota on inducing IgA immune response in T1D, suggesting that SCFAs might be potential therapeutic agents in T1D prevention and/or treatment.


Assuntos
Diabetes Mellitus Tipo 1 , Ácidos Graxos Voláteis/imunologia , Microbioma Gastrointestinal/imunologia , Imunoglobulina A/imunologia , Adolescente , Animais , Criança , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/microbiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD
19.
Mol Cell Neurosci ; 105: 103493, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32333962

RESUMO

Neuroinflammation contributes to neurodegenerative disorders, including Alzheimer's disease (AD). Gut microbes are involved in regulating systemic inflammation. Short-chain fatty acids (SCFAs), which are among the many metabolites released by gut microbes, can cross the blood-brain barrier (BBB) and interact with microglia. High concentrations of individual SCFAs decrease the inflammatory responses of peripheral monocytes; therefore, we hypothesized that SCFAs act on their own or in combinations to reduce the inflammatory response of microglia. Cultured human THP-1 monocytic cells and differentiated human HL-60 myelomonocytic cells were used to model select immune functions of human microglia. Acetate, propionate, butyrate, formate, and valerate were added to cells alone or as a mixture containing SCFAs at an approximate physiological concentration ratio. The SCFA mixture, as well as several individual SCFAs at the highest concentrations used in the mixture (15-236 µM), decreased the secretion of interleukin (IL)-1ß, monocyte chemoattractant protein (MCP)-1, tumor necrosis factor (TNF)-α, and cytotoxins by immune-stimulated THP-1 cells. GLPG 0974, a free fatty acid receptor (FFAR) 2/3 antagonist, did not block the inhibitory effect of the SCFA mixture on IL-1ß secretion by THP-1 cells while blocking the inhibitory effect of formate alone. We demonstrated that formate and valerate alone reduced the phagocytic activity of immune-stimulated THP-1 cells. Formate, but not valerate, alone also inhibited the N-formylmethionine-leucyl-phenylalanine (fMLP)-induced respiratory burst of HL-60 cells, reducing the production of reactive oxygen species (ROS). Our data indicate that SCFAs could regulate select microglial functions that are disrupted in AD.


Assuntos
Butiratos/farmacologia , Ácidos Graxos Voláteis/metabolismo , Microglia/metabolismo , Monócitos/metabolismo , Tiofenos/farmacologia , Quimiocina CCL2/metabolismo , Ácidos Graxos Voláteis/imunologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Microglia/efeitos dos fármacos , Microglia/imunologia , Monócitos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
20.
Int Immunopharmacol ; 78: 106062, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31830621

RESUMO

BACKGROUND: Alcoholic liver disease (ALD) presents one of the leading causes of cirrhosis worldwide. We have demonstrated that inulin alleviates ALD in mice. However, the exact role of hepatic macrophages in effects of inulin on ALD remains largely unclear. METHODS: In vivo, mice were divided into 4 groups: pair-fed (PF) group (PF/CON), alcohol-fed (AF) group (AF/CON), PF with inulin (INU) group (PF/INU) and AF with INU group (AF/INU). Each group was fed modified Lieber-DeCarli liquid diet with or without alcohol. In vitro, RAW264.7 cell lines were polarized to M1 macrophage (Mψ) or M2 Mψ subsets with lipopolysaccharide (LPS) or interleukin-4 (IL-4) stimulation, respectively. The effects of propionate, butyrate and valeric on macrophage M1/M2 were investigated. RESULTS: The contents of propionate, butyrate and valeric were significantly increased in AF/INU group compared with that in the AF/CON group. M1 Mψ, inducible nitric oxide synthase (iNOS) and tumor necrosis factor-α (TNF-α) in AF/INU group were significantly lower than those in AF/CON group. In contrast, M2 Mψ, arginase-1 (Arg-1), and interleukin-10 (IL-10) were notably increased in AF/INU group. In vitro, sodium propionate, sodium butyrate and sodium valerate can suppress M1 Mψ and increase M2 Mψ polarization. CONCLUSION: In ALD, inulin ameliorates the inflammation via SCFAs-inducing suppression of M1 and facilitation of M2 Mψ, which may potentially contribute to the control of the disease.


Assuntos
Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal/imunologia , Inflamação/dietoterapia , Inulina/administração & dosagem , Hepatopatias Alcoólicas/dietoterapia , Macrófagos/imunologia , Administração Oral , Animais , Técnicas de Cocultura , Modelos Animais de Doenças , Etanol/administração & dosagem , Etanol/toxicidade , Ácidos Graxos Voláteis/análise , Ácidos Graxos Voláteis/imunologia , Fezes/química , Inflamação/imunologia , Inflamação/patologia , Mucosa Intestinal/microbiologia , Fígado/citologia , Fígado/imunologia , Fígado/patologia , Hepatopatias Alcoólicas/imunologia , Hepatopatias Alcoólicas/patologia , Ativação de Macrófagos , Macrófagos/metabolismo , Masculino , Camundongos , Cultura Primária de Células , Células RAW 264.7
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA