Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.749
Filtrar
1.
Pflugers Arch ; 475(8): 945-960, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37261509

RESUMO

Proper food intake is important for maintaining good health in humans. Chocolate is known to exert anti-inflammatory effects; however, the mechanisms remain unclear. In this study, we aimed to investigate the effects of cocoa butter intake on gut immunity in rats and rabbits. Cocoa butter intake increased the lymph flow, cell density, and IL-1ß, IL-6 and IL-10 levels in mesenteric lymph. Clodronate, a macrophage depletion compound, significantly enhanced the release of all cytokines. The immunoreactivities of macrophage markers CD68 and F4/80 in the jejunal villi were significantly decreased with clodronate. Piceatannol, a selective cell surface ATP synthase inhibitor significantly reduced the cocoa butter intake-mediated releases of IL-1ß, IL-6 and IL-10. The immunoreactivities of cell surface ATP synthase were observed in rat jejunal villi. Shear stress stimulation on the myofibroblast cells isolated from rat jejunum released ATP and carbon dioxide depended with H+ release. In rabbit in vivo experiments, cocoa butter intake increased the concentrations of ATP and H+ in the portal vein. The in vitro experiments with isolated cells of rat jejunal lamina propria the pH of 3.0 and 5.0 in the medium released significantly IL-1ß and IL-6. ATP selectively released IL-10. These findings suggest that cocoa butter intake regulates the gut immunity through the release and transport of IL-1ß, IL-6, and IL-10 into mesenteric lymph vessels in a negative feedback system. In addition, the H+ and ATP released from cell surface ATP synthase in jejunal villi play key roles in the cocoa butter intake-mediated regulation of gut immunity.


Assuntos
Chocolate , Gorduras na Dieta , Trato Gastrointestinal , ATPases Translocadoras de Prótons , Animais , Ratos , Coelhos , Gorduras na Dieta/administração & dosagem , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/metabolismo , Masculino , Ratos Sprague-Dawley , Linfa/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Interleucina-10/metabolismo , Ácido Clodrônico , Jejuno/metabolismo , Resistência ao Cisalhamento , Trifosfato de Adenosina/metabolismo , Dióxido de Carbono/metabolismo , Células Cultivadas , ATPases Translocadoras de Prótons/antagonistas & inibidores , ATPases Translocadoras de Prótons/metabolismo
2.
Science ; 380(6649): 1011-1012, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37289885
3.
Science ; 380(6649): eabo2296, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37289890

RESUMO

Antibiotics (ABX) compromise the efficacy of programmed cell death protein 1 (PD-1) blockade in cancer patients, but the mechanisms underlying their immunosuppressive effects remain unknown. By inducing the down-regulation of mucosal addressin cell adhesion molecule 1 (MAdCAM-1) in the ileum, post-ABX gut recolonization by Enterocloster species drove the emigration of enterotropic α4ß7+CD4+ regulatory T 17 cells into the tumor. These deleterious ABX effects were mimicked by oral gavage of Enterocloster species, by genetic deficiency, or by antibody-mediated neutralization of MAdCAM-1 and its receptor, α4ß7 integrin. By contrast, fecal microbiota transplantation or interleukin-17A neutralization prevented ABX-induced immunosuppression. In independent lung, kidney, and bladder cancer patient cohorts, low serum levels of soluble MAdCAM-1 had a negative prognostic impact. Thus, the MAdCAM-1-α4ß7 axis constitutes an actionable gut immune checkpoint in cancer immunosurveillance.


Assuntos
Antibacterianos , Moléculas de Adesão Celular , Resistencia a Medicamentos Antineoplásicos , Microbioma Gastrointestinal , Inibidores de Checkpoint Imunológico , Tolerância Imunológica , Vigilância Imunológica , Integrinas , Mucoproteínas , Neoplasias , Animais , Humanos , Camundongos , Antibacterianos/efeitos adversos , Bactérias/imunologia , Moléculas de Adesão Celular/metabolismo , Movimento Celular , Transplante de Microbiota Fecal , Microbioma Gastrointestinal/imunologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Tolerância Imunológica/efeitos dos fármacos , Integrinas/metabolismo , Interleucina-17/metabolismo , Mucoproteínas/metabolismo , Neoplasias/imunologia , Neoplasias/terapia , Células Th17/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia
4.
Proc Natl Acad Sci U S A ; 119(41): e2209624119, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36201539

RESUMO

T cells that express the transcription factor RORγ, regulatory (Treg), or conventional (Th17) are strongly influenced by intestinal symbionts. In a genetic approach to identify mechanisms underlying this influence, we performed a screen for microbial genes implicated, in germfree mice monocolonized with Escherichia coli Nissle. The loss of capsule-synthesis genes impaired clonal expansion and differentiation of intestinal RORγ+ T cells. Mechanistic exploration revealed that the capsule-less mutants remained able to induce species-specific immunoglobulin A (IgA) and were highly IgA-coated. They could still trigger myeloid cells, and more effectively damaged epithelial cells in vitro. Unlike wild-type microbes, capsule-less mutants were mostly engulfed in intraluminal casts, large agglomerates composed of myeloid cells extravasated into the gut lumen. We speculate that sequestration in luminal casts of potentially harmful microbes, favored by IgA binding, reduces the immune system's actual exposure, preserving host-microbe equilibrium. The variable immunostimulation by microbes that has been charted in recent years may not solely be conditioned by triggering molecules or metabolites but also by physical limits to immune system exposure.


Assuntos
Trato Gastrointestinal , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Linfócitos T Reguladores , Animais , Escherichia coli , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Imunoglobulina A , Ativação Linfocitária , Camundongos , Células Mieloides , Fatores de Transcrição/metabolismo
5.
J Virol ; 96(17): e0097622, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35938870

RESUMO

Humoral immune perturbations contribute to pathogenic outcomes in persons with HIV-1 infection (PWH). Gut barrier dysfunction in PWH is associated with microbial translocation and alterations in microbial communities (dysbiosis), and IgA, the most abundant immunoglobulin (Ig) isotype in the gut, is involved in gut homeostasis by interacting with the microbiome. We determined the impact of HIV-1 infection on the antibody repertoire in the gastrointestinal tract by comparing Ig gene utilization and somatic hypermutation (SHM) in colon biopsies from PWH (n = 19) versus age and sex-matched controls (n = 13). We correlated these Ig parameters with clinical, immunological, microbiome and virological data. Gene signatures of enhanced B cell activation were accompanied by skewed frequencies of multiple Ig Variable genes in PWH. PWH showed decreased frequencies of SHM in IgA and possibly IgG, with a substantial loss of highly mutated IgA sequences. The decline in IgA SHM in PWH correlated with gut CD4+ T cell loss and inversely correlated with mucosal inflammation and microbial translocation. Diminished gut IgA SHM in PWH was driven by transversion mutations at A or T deoxynucleotides, suggesting a defect not at the AID/APOBEC3 deamination step but at later stages of IgA SHM. These results expand our understanding of humoral immune perturbations in PWH that could have important implications in understanding mucosal immune defects in individuals with chronic HIV-1 infection. IMPORTANCE The gut is a major site of early HIV-1 replication and pathogenesis. Extensive CD4+ T cell depletion in this compartment results in a compromised epithelial barrier that facilitates the translocation of microbes into the underlying lamina propria and systemic circulation, resulting in chronic immune activation. To date, the consequences of microbial translocation on the mucosal humoral immune response (or vice versa) remains poorly integrated into the panoply of mucosal immune defects in PWH. We utilized next-generation sequencing approaches to profile the Ab repertoire and ascertain frequencies of somatic hypermutation in colon biopsies from antiretroviral therapy-naive PWH versus controls. Our findings identify perturbations in the Ab repertoire of PWH that could contribute to development or maintenance of dysbiosis. Moreover, IgA mutations significantly decreased in PWH and this was associated with adverse clinical outcomes. These data may provide insight into the mechanisms underlying impaired Ab-dependent gut homeostasis during chronic HIV-1 infection.


Assuntos
Trato Gastrointestinal , Infecções por HIV , Imunoglobulina A , Hipermutação Somática de Imunoglobulina , Disbiose , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/virologia , Infecções por HIV/genética , Infecções por HIV/imunologia , HIV-1 , Humanos , Imunidade Humoral , Imunoglobulina A/genética
6.
Nature ; 607(7920): 762-768, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35794484

RESUMO

Gastrointestinal health depends on the adaptive immune system tolerating the foreign proteins in food1,2. This tolerance is paradoxical because the immune system normally attacks foreign substances by generating inflammation. Here we addressed this conundrum by using a sensitive cell enrichment method to show that polyclonal CD4+ T cells responded to food peptides, including a natural one from gliadin, by proliferating weakly in secondary lymphoid organs of the gut-liver axis owing to the action of regulatory T cells. A few food-specific T cells then differentiated into T follicular helper cells that promoted a weak antibody response. Most cells in the expanded population, however, lacked canonical T helper lineage markers and fell into five subsets dominated by naive-like or T follicular helper-like anergic cells with limited capacity to form inflammatory T helper 1 cells. Eventually, many of the T helper lineage-negative cells became regulatory T cells themselves through an interleukin-2-dependent mechanism. Our results indicate that exposure to food antigens causes cognate CD4+ naive T cells to form a complex set of noncanonical hyporesponsive T helper cell subsets that lack the inflammatory functions needed to cause gut pathology and yet have the potential to produce regulatory T cells that may suppress it.


Assuntos
Linfócitos T CD4-Positivos , Alimentos , Tolerância Imunológica , Alérgenos/imunologia , Formação de Anticorpos , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Proteínas na Dieta/imunologia , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Gliadina/imunologia , Tolerância Imunológica/imunologia , Inflamação , Interleucina-2/imunologia , Fígado/citologia , Fígado/imunologia , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Fragmentos de Peptídeos/imunologia , Células T Auxiliares Foliculares/citologia , Células T Auxiliares Foliculares/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Células Th1/citologia , Células Th1/imunologia
7.
Med Sci Monit ; 28: e935348, 2022 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-35121724

RESUMO

In recent years, immune checkpoint inhibition (ICI) therapy has made a tremendous improvement in the treatment of malignant tumors of gastrointestinal tract, especially for those with metastatic or recurrent lesions. However, while some patients benefit from ICI, others do not. In fact, predictive biomarkers can play a crucial role in screening patients who may benefit from a selected or targeted treatment, including immunotherapies such as programmed death-1/programmed death-1 ligand 1 (PD-1/PD-L1) inhibitors. A variety of techniques can be used to detect and quantify tumor biomarkers, each of which has a specific clinical application scenario and limitations. Cancer biomarkers in the gastrointestinal system involve an extremely complex network that requires careful interpretation and analysis. Different prognostic or predictive biomarkers are playing important roles in various tumor types, stages, and pathology/molecular subgroups, sometimes overlapping. Expression levels of biomarkers vary between different tumor types and even between the different lesions in the same tumor, depending on the heterogeneity of the patient, the tumor types, and the techniques of detection. The present systematic review comprehensively summarizes the potential biomarkers of immunotherapy, such as PD-1/PD-L1, total mutation burden (TMB), and tumor-infiltrating lymphocytes (TILs) in various gastrointestinal tumors, including tumors of the colon, stomach, esophagus, liver, and pancreas, to assist future application of immunotherapy and patient selection in clinical practice.


Assuntos
Inibidores de Checkpoint Imunológico/imunologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia/métodos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/imunologia , Antígeno B7-H1/imunologia , Biomarcadores Tumorais/imunologia , Trato Gastrointestinal/imunologia , Humanos , Linfócitos do Interstício Tumoral/imunologia
8.
Gut Microbes ; 14(1): 2035659, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35167406

RESUMO

Gut microbiota and its association with cancer development/treatment has been intensively studied during the past several years. Currently, there is a growing interest toward next-generation probiotics (NGPs) as therapeutic agents that alter gut microbiota and impact on cancer development. In the present review we focus on three emerging NGPs, namely Faecalibacterium prausnitzii, Akkermansia muciniphila, and Bacteroides fragilis as their presence in the digestive tract can have an impact on cancer incidence. These NGPs enhance gastrointestinal immunity, maintain intestinal barrier integrity, produce beneficial metabolites, act against pathogens, improve immunotherapy efficacy, and reduce complications associated with chemotherapy and radiotherapy. Notably, the use of NGPs in cancer patients does not have a long history and, although their safety remains relatively undefined, recently published data has shown that they are non-toxigenic. Notwithstanding, A. muciniphila may promote colitis whereas enterotoxigenic B. fragilis stimulates chronic inflammation and participates in colorectal carcinogenesis. Nevertheless, the majority of B. fragilis strains provide a beneficial effect to the host, are non-toxigenic and considered as the best current NGP candidate. Overall, emerging studies indicate a beneficial role of these NGPs in the prevention of carcinogenesis and open new promising therapeutic options for cancer patients.


Assuntos
Microbioma Gastrointestinal , Neoplasias/tratamento farmacológico , Probióticos/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Terapia Combinada , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Humanos , Neoplasias/imunologia , Neoplasias/microbiologia , Neoplasias/radioterapia
9.
J Exp Med ; 219(3)2022 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-35195681

RESUMO

The gastrointestinal tract contains trillions of microorganisms that exist symbiotically with the host due to a tolerant, regulatory cell-rich intestinal immune system. However, this intimate relationship with the microbiome inevitably comes with risks, with intestinal organisms being the most common cause of bacteremia. The vasculature of the brain-lining meninges contains fenestrated endothelium, conferring vulnerability to invasion by circulating microbes. We propose that this has evolutionarily led to close links between gut and meningeal immunity, to prime the central nervous system defense against the most likely invaders. This paradigm is exemplified by the dural venous sinus IgA defense system, where the antibody repertoire mirrors that of the gut.


Assuntos
Trato Gastrointestinal/imunologia , Meninges/imunologia , Animais , Microbioma Gastrointestinal/imunologia , Humanos , Imunoglobulina A/imunologia , Meninges/microbiologia , Modelos Imunológicos , Plasmócitos/imunologia
10.
Nutrients ; 14(2)2022 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-35057455

RESUMO

Coronavirus disease 2019 (COVID-19) was declared a pandemic at the beginning of 2020, causing millions of deaths worldwide. Millions of vaccine doses have been administered worldwide; however, outbreaks continue. Probiotics are known to restore a stable gut microbiota by regulating innate and adaptive immunity within the gut, demonstrating the possibility that they may be used to combat COVID-19 because of several pieces of evidence suggesting that COVID-19 has an adverse impact on gut microbiota dysbiosis. Thus, probiotics and their metabolites with known antiviral properties may be used as an adjunctive treatment to combat COVID-19. Several clinical trials have revealed the efficacy of probiotics and their metabolites in treating patients with SARS-CoV-2. However, its molecular mechanism has not been unraveled. The availability of abundant data resources and computational methods has significantly changed research finding molecular insights between probiotics and COVID-19. This review highlights computational approaches involving microbiome-based approaches and ensemble-driven docking approaches, as well as a case study proving the effects of probiotic metabolites on SARS-CoV-2.


Assuntos
COVID-19/microbiologia , COVID-19/terapia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Probióticos/farmacologia , Probióticos/uso terapêutico , Computadores Moleculares , Disbiose/tratamento farmacológico , Feminino , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Humanos , Masculino , Simulação de Acoplamento Molecular , Probióticos/metabolismo
11.
Pharmacol Res ; 175: 106025, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34883211

RESUMO

Originating from Eastern Asia, the plant Cannabis sativa has been used for centuries as a medicinal treatment. The unwanted psychotropic effects of one of its major components, Δ9-tetrahydrocannabinol, discouraged its therapeutic employment until, recently, the discovery of cannabinoids receptors and their endogenous ligands endocannabinoids reignited the interest. The endocannabinoid system has lately been found to play an important role in the maintenance of human health, both centrally and peripherally. However, the initial idea of the endocannabinoid system structure has been quickly understood to be too simplistic and, as new receptors, mediators, and enzymes have been discovered to participate in a complex relationship, the new, more comprehensive term "expanded endocannabinoid system" or "endocannabinoidome", has taken over. The discovery of other endocannabinoid-like receptors, such as the G protein-coupled receptor 119 and G protein-coupled receptor 55, has opened the way to the development of potential therapeutic targets for the treatment of various metabolic disorders. In addition, recent findings have also provided evidence suggesting the potential therapeutic link between the endocannabinoidome and various inflammatory-based gut diseases, such as inflammatory bowel disease and cancer. This review will provide an introduction to the endocannabinoidome, focusing on its modulatory role in the gastrointestinal tract and on the interest generated by the link between gut microbiota, the endocannabinoid system and metabolic diseases such as inflammatory bowel disease, type-2 diabetes and obesity. In addition, we will look at the potential novel aspects and benefits of drugs targeting the endocannabinoid system.


Assuntos
Endocanabinoides/metabolismo , Trato Gastrointestinal/metabolismo , Tecido Adiposo/metabolismo , Animais , Eixo Encéfalo-Intestino , Diabetes Mellitus Tipo 2/metabolismo , Endocanabinoides/imunologia , Neoplasias Gastrointestinais/metabolismo , Trato Gastrointestinal/imunologia , Homeostase , Humanos , Imunidade nas Mucosas , Doenças Inflamatórias Intestinais/metabolismo , Obesidade/metabolismo , Receptores de Canabinoides/metabolismo , Termogênese
12.
J Nutr Biochem ; 101: 108926, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34848335

RESUMO

Short-chain fatty acids (SCFAs) play a pivotal role in maintaining intestinal homeostasis. We aimed to investigate the effects of SCFA supplementation on gut inflammation and microbiota composition in a murine colitis model. Mice were fed with sodium butyrate or a mixture of SCFAs in the drinking water for 2 weeks, followed by 2% dextran sulfate sodium (DSS) for 7 d. After euthanasia, mouse colons were extracted to examine histological findings. Flow cytometry of the mouse colon tissues was performed to assess T cell differentiation. Changes in gut microbiota were assessed by high-throughput sequencing of the mouse feces. There were no significant differences in weight change, colonic length, or histologic inflammation score between the DSS, butyrate, and SCFA mix groups. However, flow cytometry revealed that both the expression of CD4+Foxp3+ regulatory T cells and of IL-17-producing T cells were increased in the butyrate and SCFA mix groups. Microbial compositions of the butyrate and SCFA mix groups were significantly different from those of the control and DSS groups in principal coordinate analysis. Relative abundances of the phyla Verrucomicrobia and Proteobacteria, species Akkermansia muciniphila and Escherichia fergusonii were increased in the butyrate and SCFA mix groups. Genera Roseburia and Lactobacillus showed a negative correlation with the degree of colitis, whereas genera Escherichia and Mucispirillum showed a positive correlation. SCFA supplementation did not result in a significant reduction in colon inflammation, but it promoted both regulatory T cell and IL-17-producing T cell expression, and increased both protective and aggressive gut microbiota.


Assuntos
Butiratos/administração & dosagem , Suplementos Nutricionais , Ácidos Graxos Voláteis/administração & dosagem , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Animais , Diferenciação Celular , Colite/imunologia , Colite/microbiologia , Colite/patologia , Colo/patologia , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/patologia , Doenças Inflamatórias Intestinais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia
13.
Hepatology ; 75(3): 518-530, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34633679

RESUMO

BACKGROUND AND AIMS: The "gut homing" hypothesis suggests the pathogenesis of primary sclerosing cholangitis (PSC) is driven by aberrant hepatic expression of gut adhesion molecules and subsequent recruitment of gut-derived T cells to the liver. However, inconsistencies lie within this theory including an absence of investigations and comparisons with other chronic liver diseases (CLD). Here, we examine "the gut homing theory" in patients with PSC with associated inflammatory bowel disease (PSC-IBD) and across multiple inflammatory liver diseases. APPROACH AND RESULTS: Expression of MAdCAM-1, CCL25, and E-Cadherin were assessed histologically and using RT-PCR on explanted liver tissue from patients with CLD undergoing OLT and in normal liver. Liver mononuclear cells were isolated from explanted tissue samples and the expression of gut homing integrins and cytokines on hepatic infiltrating gut-derived T cells was assessed using flow cytometry. Hepatic expression of MAdCAM-1, CCL25 and E-Cadherin was up-regulated in all CLDs compared with normal liver. There were no differences between disease groups. Frequencies of α4ß7, αEß7, CCR9, and GPR15 expressing hepatic T cells was increased in PSC-IBD, but also in CLD controls, compared with normal liver. ß7 expressing hepatic T cells displayed an increased inflammatory phenotype compared with ß7 negative cells, although this inflammatory cytokine profile was present in both the inflamed and normal liver. CONCLUSIONS: These findings refute the widely accepted "gut homing" hypothesis as the primary driver of PSC and indicate that aberrant hepatic recruitment of gut-derived T cells is not unique to PSC, but is a panetiological feature of CLD.


Assuntos
Antígenos CD/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Caderinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Quimiocinas CC/metabolismo , Colangite Esclerosante , Trato Gastrointestinal , Hepatopatias , Fígado , Mucoproteínas/metabolismo , Moléculas de Adesão Celular/isolamento & purificação , Colangite Esclerosante/imunologia , Colangite Esclerosante/metabolismo , Colangite Esclerosante/patologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/patologia , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Cadeias beta de Integrinas/metabolismo , Fígado/metabolismo , Fígado/patologia , Hepatopatias/classificação , Hepatopatias/metabolismo , Hepatopatias/patologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo
14.
Front Immunol ; 12: 778098, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925352

RESUMO

The gut microbiota is a complex group of microorganisms that is not only closely related to intestinal immunity but also affects the whole immune system of the body. Antimicrobial peptides and reactive oxygen species participate in the regulation of gut microbiota homeostasis in invertebrates. However, it is unclear whether nitric oxide, as a key mediator of immunity that plays important roles in antipathogen activity and immune regulation, participates in the regulation of gut microbiota homeostasis. In this study, we identified a nitric oxide synthase responsible for NO production in the shrimp Marsupenaeus japonicus. The expression of Nos and the NO concentration in the gastrointestinal tract were increased significantly in shrimp orally infected with Vibrio anguillarum. After RNA interference of Nos or treatment with an inhibitor of NOS, L-NMMA, NO production decreased and the gut bacterial load increased significantly in shrimp. Treatment with the NO donor, sodium nitroprusside, increased the NO level and reduced the bacterial load significantly in the shrimp gastrointestinal tract. Mechanistically, V. anguillarum infection increased NO level via upregulation of NOS and induced phosphorylation of ERK. The activated ERK phosphorylated the NF-κB-like transcription factor, dorsal, and caused nuclear translocation of dorsal to increase expression of antimicrobial peptides (AMPs) responsible for bacterial clearance. In summary, as a signaling molecule, NOS-produced NO regulates intestinal microbiota homeostasis by promoting AMP expression against infected pathogens via the ERK-dorsal pathway in shrimp.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , NF-kappa B/metabolismo , Óxido Nítrico Sintase/metabolismo , Penaeidae/microbiologia , Vibrioses/microbiologia , Vibrio/patogenicidade , Animais , Peptídeos Antimicrobianos/metabolismo , Carga Bacteriana , Trato Gastrointestinal/enzimologia , Trato Gastrointestinal/imunologia , Homeostase , Óxido Nítrico/metabolismo , Penaeidae/enzimologia , Penaeidae/imunologia , Fosforilação , Transdução de Sinais , Vibrio/imunologia , Vibrioses/enzimologia , Vibrioses/imunologia
15.
Front Immunol ; 12: 741233, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867963

RESUMO

The gut microbiota is an important regulator for maintaining the organ microenvironment through effects on the gut-vital organs axis. Respiratory tract infections are one of the most widespread and harmful diseases, especially in the last 2 years. Many lines of evidence indicate that the gut microbiota and its metabolites can be considered in therapeutic strategies to effectively prevent and treat respiratory diseases. However, due to the different gut microbiota composition in children compared to adults and the dynamic development of the immature immune system, studies on the interaction between children's intestinal flora and respiratory infections are still lacking. Here, we describe the changes in the gut microbiota of children with respiratory tract infections and explain the relationship between the microbiota of children with their immune function and disease development. In addition, we will provide perspectives on the direct manipulation of intestinal microbes to prevent or treat pediatric respiratory infections.


Assuntos
Microbioma Gastrointestinal/imunologia , Infecções Respiratórias/microbiologia , Animais , Criança , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Humanos , Pulmão/imunologia , Pulmão/microbiologia , Infecções Respiratórias/terapia
16.
Front Immunol ; 12: 766586, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34868019

RESUMO

Cytokines are powerful mediators of inflammation. Consequently, their potency is regulated in many ways to protect the host. Several cytokines, including IL-22, have coordinating binding proteins or soluble receptors that bind to the cytokine, block the interaction with the cellular receptor, and thus prevent cellular signaling. IL-22 is a critical cytokine in the modulation of tissue responses during inflammation and is highly upregulated in many chronic inflammatory disease patients, including those with psoriasis, rheumatoid arthritis, and inflammatory bowel disease (IBD). In healthy individuals, low levels of IL-22 are secreted by immune cells, mainly in the gastrointestinal (GI) tract. However, much of this IL-22 is likely not biologically active due to the high levels of IL-22 binding protein (IL-22BP) produced by intestinal dendritic cells (DCs). IL-22BP is a soluble receptor homolog that binds to IL-22 with greater affinity than the membrane spanning receptor. Much is known regarding the regulation and function of IL-22 in health and disease. However, less is known about IL-22BP. In this review, we will focus on IL-22BP, including its regulation, role in IL-22 biology and inflammation, and promise as a therapeutic. IL-22 can be protective or pathogenic, depending on the context of inflammation. IL-22BP also has divergent roles. Ongoing and forthcoming studies will expand our knowledge of IL-22BP and IL-22 biology, and suggest that IL-22BP holds promise as a way to regulate IL-22 biology in patients with chronic inflammatory disease.


Assuntos
Interleucinas/fisiologia , Receptores de Interleucina/fisiologia , Trato Gastrointestinal/imunologia , Humanos , Inflamação/imunologia , Fígado/imunologia , Receptores de Interleucina/análise , Pele/imunologia
17.
Science ; 374(6571): 1087-1092, 2021 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-34822299

RESUMO

The brain and gastrointestinal tract are critical sensory organs responsible for detecting, relaying, integrating, and responding to signals derived from the internal and external environment. At the interface of this sensory function, immune cells in the intestines and brain consistently survey environmental factors, eliciting responses that inform on the physiological state of the body. Recent research reveals that cross-talk along the gut-brain axis regulates inflammatory nociception, inflammatory responses, and immune homeostasis. Here, we discuss molecular and cellular mechanisms involved in the signaling of inflammation across the gut-brain axis. We further highlight interactions between the gut and the brain in inflammation-associated diseases.


Assuntos
Eixo Encéfalo-Intestino/fisiologia , Encéfalo/fisiologia , Trato Gastrointestinal/fisiologia , Inflamação/fisiopatologia , Transdução de Sinais , Animais , Encéfalo/imunologia , Eixo Encéfalo-Intestino/imunologia , Trato Gastrointestinal/imunologia , Humanos , Inflamação/imunologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/fisiopatologia , Neurônios/fisiologia , Nociceptividade
18.
PLoS One ; 16(11): e0260012, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34793546

RESUMO

INTRODUCTION: Study of inflammatory cytokines in patients with caustic gastrointestinal tract injury is sketchy. This study investigated the cytokine profiling of patients with caustic substance ingestion, and analyzed the differences between patients with severe and mild injury. METHODS: This prospective, cross-sectional study enrolled 22 patients admitted to Chang Gung Memorial Hospital between March and October 2018. All patients underwent esophagogastroduodenoscopy in 24 hours. Patients were categorized into two subgroups, as mild (<2b, n = 11) or severe (≥2b, n = 11) group. RESULTS: The neutrophil count was higher in severe than mild group (P = 0.032). Patients in mild and severe groups exhibited significantly higher circulating inflammatory cytokines than healthy control, including interleukin (IL)-2, IL-5, IL-8, IL-9, IL-12, IL-13, interferon-gamma inducible protein-10, macrophage inflammatory protein-1 beta, regulated upon activation, normal T cell expressed and presumably secreted and tumor necrosis factor-alpha. Furthermore, the levels of IL-2 and tumor necrosis factor-alpha were significantly higher in patients with severe group than mild group. Although there was no difference in cumulative survival between both groups (P = 0.147), the severe group received more operations (P = 0.035) and suffered more gastrointestinal complications (P = 0.035) than mild group. CONCLUSION: Caustic substance ingestion produces mucosal damages and leads to excessive neutrophils and inflammatory cytokines in peripheral blood.


Assuntos
Citocinas/análise , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/lesões , Traumatismos Abdominais , Adulto , Idoso , Idoso de 80 Anos ou mais , Queimaduras Químicas/genética , Queimaduras Químicas/imunologia , Cáusticos/toxicidade , Estudos Transversais , Citocinas/sangue , Feminino , Humanos , Interleucinas/análise , Interleucinas/sangue , Contagem de Leucócitos/métodos , Masculino , Pessoa de Meia-Idade , Neutrófilos/metabolismo , Estudos Prospectivos , Taiwan , Traumatismos Torácicos , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/sangue
19.
Nutrients ; 13(11)2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34836408

RESUMO

The prevalence of asthma is increasing, but the cause remains under debate. Research currently focuses on environmental and dietary factors that may impact the gut-lung axis. Dietary fibers are considered to play a crucial role in supporting diversity and activity of the microbiome, as well as immune homeostasis in the gut and lung. This review discusses the current state of knowledge on how dietary fibers and their bacterial fermentation products may affect the pathophysiology of allergic asthma. Moreover, the impact of dietary fibers on early type 2 asthma management, as shown in both pre-clinical and clinical studies, is described. Short-chain fatty acids, fiber metabolites, modulate host immunity and might reduce the risk of allergic asthma development. Underlying mechanisms include G protein-coupled receptor activation and histone deacetylase inhibition. These results are supported by studies in mice, children and adults with allergic asthma. Fibers might also exert direct effects on the immune system via yet to be elucidated mechanisms. However, the effects of specific types of fiber, dosages, duration of treatment, and combination with probiotics, need to be explored. There is an urgent need to further valorize the potential of specific dietary fibers in prevention and treatment of allergic asthma by conducting more large-scale dietary intervention trials.


Assuntos
Asma/imunologia , Fibras na Dieta/metabolismo , Trato Gastrointestinal/imunologia , Hipersensibilidade/imunologia , Pulmão/imunologia , Animais , Asma/microbiologia , Fibras na Dieta/uso terapêutico , Trato Gastrointestinal/microbiologia , Humanos , Hipersensibilidade/microbiologia , Pulmão/microbiologia , Camundongos , Microbiota/imunologia
20.
Cells ; 10(11)2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34831429

RESUMO

The Farnesoid-X Receptor, FXR, is a nuclear bile acid receptor. Its originally described function is in bile acid synthesis and regulation within the liver. More recently, however, FXR has been increasingly appreciated for its breadth of function and expression across multiple organ systems, including the intestine. While FXR's role within the liver continues to be investigated, increasing literature indicates that FXR has important roles in responding to inflammation, maintaining intestinal epithelial barrier function, and regulating immunity within the gastrointestinal (GI) tract. Given the complicated and multi-factorial nature of intestinal barrier dysfunction, it is not surprising that FXR's role appears equally complicated and not without conflicting data in different model systems. Recent work has suggested translational applications of FXR modulation in GI pathology; however, a better understanding of FXR physiology is necessary for these treatments to gain widespread use in human disease. This review aims to discuss current scientific work on the role of FXR within the GI tract, specifically in its role in intestinal inflammation, barrier function, and immune response, while also exploring areas of controversy.


Assuntos
Trato Gastrointestinal/imunologia , Trato Gastrointestinal/fisiopatologia , Imunidade Inata , Receptores Citoplasmáticos e Nucleares/metabolismo , Trato Gastrointestinal/patologia , Humanos , Inflamação/patologia , Modelos Biológicos , Junções Íntimas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...