Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Nat Rev Immunol ; 24(8): 577-595, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38565643

RESUMEN

The short-chain fatty acids (SCFAs) butyrate, propionate and acetate are microbial metabolites and their availability in the gut and other organs is determined by environmental factors, such as diet and use of antibiotics, that shape the diversity and metabolism of the microbiota. SCFAs regulate epithelial barrier function as well as mucosal and systemic immunity via evolutionary conserved processes that involve G protein-coupled receptor signalling or histone deacetylase activity. Indicatively, the anti-inflammatory role of butyrate is mediated through direct effects on the differentiation of intestinal epithelial cells, phagocytes, B cells and plasma cells, and regulatory and effector T cells. Intestinally derived SCFAs also directly and indirectly affect immunity at extra-intestinal sites, such as the liver, the lungs, the reproductive tract and the brain, and have been implicated in a range of disorders, including infections, intestinal inflammation, autoimmunity, food allergies, asthma and responses to cancer therapies. An ecological understanding of microbial communities and their interrelated metabolic states, as well as the engineering of butyrogenic bacteria may support SCFA-focused interventions for the prevention and treatment of immune-mediated diseases.


Asunto(s)
Dieta , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Humanos , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/inmunología , Animales , Microbioma Gastrointestinal/inmunología , Microbiota/inmunología , Butiratos/metabolismo
2.
Inflammation ; 45(1): 372-386, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34792688

RESUMEN

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.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Colitis/metabolismo , Colon/metabolismo , Fosfatasa 6 de Especificidad Dual/metabolismo , Ácidos Grasos Volátiles/metabolismo , Mucosa Intestinal/metabolismo , MicroARNs/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/inmunología , Colitis/inmunología , Colitis/patología , Colon/inmunología , Colon/patología , Fosfatasa 6 de Especificidad Dual/inmunología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Ácidos Grasos Volátiles/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/inmunología
3.
Int J Mol Sci ; 22(16)2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34445184

RESUMEN

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.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Pulmón/microbiología , Inmunidad Adaptativa , Animales , Aspergilosis/microbiología , Aspergillus fumigatus/fisiología , Ácidos Grasos Volátiles/inmunología , Interacciones Huésped-Patógeno , Inmunidad Innata , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Microbiota
4.
Biomed Pharmacother ; 140: 111542, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34088571

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Flavonas/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Melanoma/inmunología , Melanoma/terapia , Microbiota/efectos de los fármacos , Umbeliferonas/farmacología , Animales , Ciego/microbiología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ácidos Grasos Volátiles/inmunología , Heces/microbiología , Femenino , Inmunoterapia/métodos , Indoles/farmacología , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
5.
Infect Immun ; 89(9): e0018821, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34097474

RESUMEN

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.


Asunto(s)
Antiinfecciosos/uso terapéutico , Ácidos Grasos Volátiles/uso terapéutico , Infecciones/tratamiento farmacológico , Pulmón/efectos de los fármacos , Animales , Antiinfecciosos/inmunología , Antiinfecciosos/metabolismo , Ácidos Grasos Volátiles/inmunología , Ácidos Grasos Volátiles/metabolismo , Humanos , Infecciones/inmunología , Infecciones/microbiología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/virología , Viabilidad Microbiana , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/virología , Transducción de Señal/inmunología , Virulencia
6.
Biomed Pharmacother ; 141: 111817, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34126349

RESUMEN

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.


Asunto(s)
Infecciones Bacterianas/inmunología , Ácidos Grasos Volátiles/farmacología , Factores Inmunológicos/farmacología , Microbiota , Animales , Ácidos Grasos Volátiles/inmunología , Microbioma Gastrointestinal , Humanos
7.
Gut Microbes ; 13(1): 1-22, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33590776

RESUMEN

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.


Asunto(s)
Bacterias/metabolismo , Microbioma Gastrointestinal/fisiología , Enfermedades no Transmisibles , Transducción de Señal/inmunología , Amidas/inmunología , Amidas/metabolismo , Bacterias/clasificación , Bacterias/aislamiento & purificación , Ácidos y Sales Biliares/inmunología , Ácidos y Sales Biliares/metabolismo , Colina/inmunología , Colina/metabolismo , Susceptibilidad a Enfermedades/inmunología , Susceptibilidad a Enfermedades/microbiología , Ácidos Grasos Volátiles/inmunología , Ácidos Grasos Volátiles/metabolismo , Humanos , Sistema Inmunológico/inmunología , Indoles/inmunología , Indoles/metabolismo , Poliaminas/inmunología , Poliaminas/metabolismo , Vitaminas/inmunología , Vitaminas/metabolismo
8.
Cells ; 10(1)2021 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-33435197

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales/inmunología , Ácidos Grasos Volátiles/inmunología , Herpesvirus Humano 4/inmunología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/virología , Vitamina D/metabolismo , Adulto , Estudios de Casos y Controles , Ambiente , Femenino , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Masculino , Persona de Mediana Edad , Vitamina D/análogos & derivados
9.
Front Immunol ; 12: 761820, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069532

RESUMEN

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.


Asunto(s)
Alimentación Animal , Carpas , Ácidos Grasos Volátiles/inmunología , Microbioma Gastrointestinal , Inmunomodulación/efectos de los fármacos , beta-Glucanos/farmacología , Animales , Carpas/inmunología , Carpas/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología
10.
J Infect Dis ; 223(12 Suppl 2): S194-S200, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-33326565
11.
Acta Biochim Pol ; 67(4): 485-493, 2020 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-33332076

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Colitis/tratamiento farmacológico , Colon/inmunología , Microbioma Gastrointestinal/inmunología , Lactobacillus plantarum/química , Polisacáridos Bacterianos/farmacología , Animales , Antiinflamatorios/aislamiento & purificación , Peso Corporal/efectos de los fármacos , Clostridiales , Colitis/inducido químicamente , Colitis/inmunología , Colitis/microbiología , Colon/metabolismo , Colon/microbiología , Sulfato de Dextran , Ácidos Grasos Volátiles/inmunología , Ácidos Grasos Volátiles/metabolismo , Heces/microbiología , Expresión Génica , Inmunidad Innata , Interferón gamma/genética , Interferón gamma/inmunología , Interleucinas/genética , Interleucinas/inmunología , Masculino , Ratones , Ratones Endogámicos ICR , Polisacáridos Bacterianos/aislamiento & purificación , Prednisolona/análogos & derivados , Prednisolona/farmacología , ARN Ribosómico 16S/genética , Ruminococcus , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
12.
Front Immunol ; 11: 590685, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33363537

RESUMEN

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.


Asunto(s)
Carcinogénesis/inmunología , Diabetes Mellitus/inmunología , Ácidos Grasos Volátiles/inmunología , Inflamación/inmunología , Animales , Diabetes Mellitus/microbiología , Disbiosis/inmunología , Disbiosis/microbiología , Microbioma Gastrointestinal , Humanos , Inflamación/microbiología , Neoplasias/inmunología , Neoplasias/microbiología
13.
Int J Mol Sci ; 21(22)2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33227973

RESUMEN

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.


Asunto(s)
Aterosclerosis/inmunología , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Innata , Inmunomodulación , Animales , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/microbiología , Aterosclerosis/patología , Ensayos Clínicos como Asunto , Citocinas/inmunología , Citocinas/metabolismo , Progresión de la Enfermedad , Disbiosis/tratamiento farmacológico , Disbiosis/microbiología , Disbiosis/patología , Ácidos Grasos Volátiles/inmunología , Ácidos Grasos Volátiles/metabolismo , Humanos , Factores Inmunológicos/uso terapéutico , Inflamasomas/inmunología , Inflamasomas/metabolismo , Inflamación , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Metilaminas/inmunología , Metilaminas/metabolismo
14.
Nat Commun ; 11(1): 4457, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32901017

RESUMEN

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.


Asunto(s)
Ácidos Grasos Volátiles/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Innata , Interleucinas/biosíntesis , Animales , Butiratos/inmunología , Butiratos/metabolismo , Butiratos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Citrobacter rodentium , Colitis/inmunología , Colitis/microbiología , Colitis/prevención & control , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/prevención & control , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/farmacología , Microbioma Gastrointestinal/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Técnicas In Vitro , Interleucinas/deficiencia , Interleucinas/genética , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Regiones Promotoras Genéticas , Receptores de Hidrocarburo de Aril/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Interleucina-22
15.
Front Immunol ; 11: 1427, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32903327

RESUMEN

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.


Asunto(s)
Ácidos Grasos Volátiles , Hipersensibilidad , Leche Humana/química , Adulto , Ácidos Grasos Volátiles/inmunología , Femenino , Humanos , Hipersensibilidad/inmunología , Lactante , Leche Humana/metabolismo , Madres
16.
Int J Mol Sci ; 21(14)2020 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-32664466

RESUMEN

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.


Asunto(s)
Disbiosis/complicaciones , Células Epiteliales/efectos de los fármacos , Ácidos Grasos Volátiles/efectos adversos , Mucosa Bucal/microbiología , Periodontitis/etiología , Apoptosis/efectos de los fármacos , Biopelículas , Butiratos/farmacología , Adhesión Celular/efectos de los fármacos , Forma de la Célula , Disbiosis/microbiología , Ácidos Grasos Volátiles/inmunología , Humanos , Microbiota , Mucosa Bucal/citología , Bolsa Periodontal/microbiología , Periodontitis/tratamiento farmacológico , Bicarbonato de Sodio/farmacología , Bicarbonato de Sodio/uso terapéutico
17.
J Am Soc Nephrol ; 31(7): 1445-1461, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32482686

RESUMEN

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.


Asunto(s)
Fibras de la Dieta/administración & dosificación , Ácidos Grasos Volátiles/inmunología , Microbioma Gastrointestinal/inmunología , Rechazo de Injerto/prevención & control , Tolerancia Inmunológica/efectos de los fármacos , Linfocitos T Reguladores , Enfermedad Aguda , Aloinjertos/inmunología , Animales , Ácido Butírico/farmacología , Enfermedad Crónica , Suplementos Dietéticos , Disbiosis/etiología , Disbiosis/microbiología , Disbiosis/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Rechazo de Injerto/patología , Rechazo de Injerto/fisiopatología , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Trasplante de Riñón/efectos adversos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Acetato de Sodio/farmacología
18.
Artículo en Inglés | MEDLINE | ID: mdl-32413493

RESUMEN

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.


Asunto(s)
Quitosano/administración & dosificación , Ácidos Grasos Volátiles/inmunología , Penaeidae/inmunología , Probióticos/administración & dosificación , Amoníaco/toxicidad , Alimentación Animal , Animales , Suplementos Dietéticos , Ácidos Grasos Volátiles/metabolismo , Inmunidad Innata/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Intestinos/inmunología , Penaeidae/efectos de los fármacos , Penaeidae/crecimiento & desarrollo , Penaeidae/metabolismo
19.
JCI Insight ; 5(10)2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32298241

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 1 , Ácidos Grasos Volátiles/inmunología , Microbioma Gastrointestinal/inmunología , Inmunoglobulina A/inmunología , Adolescente , Animales , Niño , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/microbiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos NOD
20.
Mol Cell Neurosci ; 105: 103493, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32333962

RESUMEN

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.


Asunto(s)
Butiratos/farmacología , Ácidos Grasos Volátiles/metabolismo , Microglía/metabolismo , Monocitos/metabolismo , Tiofenos/farmacología , Quimiocina CCL2/metabolismo , Ácidos Grasos Volátiles/inmunología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Microglía/efectos de los fármacos , Microglía/inmunología , Monocitos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA