Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Immunity ; 49(5): 915-928.e5, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30446384

RESUMO

Innate lymphoid cells (ILCs) are important for mucosal immunity. The intestine harbors all ILC subsets, but how these cells are balanced to achieve immune homeostasis and mount appropriate responses during infection remains elusive. Here, we show that aryl hydrocarbon receptor (Ahr) expression in the gut regulates ILC balance. Among ILCs, Ahr is most highly expressed by gut ILC2s and controls chromatin accessibility at the Ahr locus via positive feedback. Ahr signaling suppresses Gfi1 transcription-factor-mediated expression of the interleukin-33 (IL-33) receptor ST2 in ILC2s and expression of ILC2 effector molecules IL-5, IL-13, and amphiregulin in a cell-intrinsic manner. Ablation of Ahr enhances anti-helminth immunity in the gut, whereas genetic or pharmacological activation of Ahr suppresses ILC2 function but enhances ILC3 maintenance to protect the host from Citrobacter rodentium infection. Thus, the host regulates the gut ILC2-ILC3 balance by engaging the Ahr pathway to mount appropriate immunity against various pathogens.


Assuntos
Imunidade Inata , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Biomarcadores , Cromatina/genética , Cromatina/metabolismo , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/microbiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Loci Gênicos , Interações Hospedeiro-Parasita/imunologia , Imunidade nas Mucosas/genética , Imunofenotipagem , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , Receptores de Hidrocarboneto Arílico/genética , Transcriptoma
2.
Mol Microbiol ; 2024 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-38703041

RESUMO

The human digestive system harbors a vast diversity of commensal bacteria and maintains a symbiotic relationship with them. However, imbalances in the gut microbiota accompany various diseases, such as inflammatory bowel diseases (IBDs) and colorectal cancers (CRCs), which significantly impact the well-being of populations globally. Glycosylation of the mucus layer is a crucial factor that plays a critical role in maintaining the homeostatic environment in the gut. This review delves into how the gut microbiota, immune cells, and gut mucus layer work together to establish a balanced gut environment. Specifically, the role of glycosylation in regulating immune cell responses and mucus metabolism in this process is examined.

3.
Crit Rev Food Sci Nutr ; 63(19): 3895-3911, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-34748438

RESUMO

ß-glucan from cereals such as wheat, barley, oats and rye are a water-soluble dietary fiber, which are composed of repeating (1→4)-ß-bond ß-D-glucopyranosyl units and a single (1→3)-ß-D-bond separated unit. ß-glucan has a series of physicochemical properties (such as viscosity, gelling properties, solubility, etc.), which can be used as a food gel and fat substitute. Its structure endows the healthy functions, including anti-oxidative stress, lowering blood glucose and serum cholesterol, regulating metabolic syndrome and exerting gut immunity via gut microbiota. Due to their unique structural properties and efficacy, cereal ß-glucan are not only applied in food substrates in the food industry, but also in food coatings and packaging. This article reviewed the applications of cereal ß-glucan in hydrogels, aerogels, intelligent packaging systems and targeted delivery carriers in recent years. Cereal ß-glucan in edible film and gel packaging applications are becoming more diversified and intelligent in recent years. Those advances provide a potential solution based on cereal ß-glucan as biodegradable substances for immune regulation delivery system and intelligent gelling material in the biomedicine field.


Assuntos
Microbioma Gastrointestinal , beta-Glucanas , Grão Comestível/química , Glicemia/metabolismo , Solubilidade , Avena/química , Fibras na Dieta/análise
4.
Dev Psychobiol ; 65(7): e22424, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37860905

RESUMO

Prenatal maternal stress (PNMS)-characterized by exposure to stress, anxiety, depression, or intimate partner violence-has been linked to biological alterations in infants, including disruptions to their intestinal microbiota, which have long-term implications for children's developmental outcomes. Significant research has been done examining the effects of PNMS on the microbiome in animals, but less is known about these effects in human research. The current systematic review aimed to synthesize current findings on the association between PNMS and mother and infant microbiomes. Medline, Embase, PsycInfo, Web of Science, and Eric databases were searched through to February 2022. A total of eight studies (n = 2219 infants, 2202 mothers) were included in the qualitative synthesis. Findings provided promising evidence of the role that PNMS plays in altering the microbial composition, diversity, and gut immunity in mothers and infants. Notably, majority of included studies found that higher PNMS was linked to increases in genera from the phylum Proteobacteria. The factors influencing these effects are explored including nutrition, birth mode, and parenting behaviors. Potential interventions to mitigate the adverse effects of PNMS are discussed, along with recommendations for future studies with longitudinal designs to better understand the appropriate type and timing of interventions needed to promote "healthy" maternal and infant microbial functioning.


Assuntos
Microbioma Gastrointestinal , Mães , Feminino , Criança , Gravidez , Animais , Humanos , Lactente , Estresse Psicológico/microbiologia , Ansiedade , Transtornos de Ansiedade
5.
Int J Mol Sci ; 24(19)2023 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-37834203

RESUMO

The bidirectional communication between the gut and central nervous system (CNS) through microbiota is known as the microbiota-gut-brain axis. The brain, through the enteric neural innervation and the vagus nerve, influences the gut physiological activities (motility, mucin, and peptide secretion), as well as the development of the mucosal immune system. Conversely, the gut can influence the CNS via intestinal microbiota, its metabolites, and gut-homing immune cells. Growing evidence suggests that gut immunity is critically involved in gut-brain communication during health and diseases, including multiple sclerosis (MS). The gut microbiota can influence the development and function of gut immunity, and conversely, the innate and adaptive mucosal immunity can influence microbiota composition. Gut and systemic immunity, along with gut microbiota, are perturbed in MS. Diet and disease-modifying therapies (DMTs) can affect the composition of the gut microbial community, leading to changes in gut and peripheral immunity, which ultimately affects MS. A high-fat diet is highly associated with gut dysbiosis-mediated inflammation and intestinal permeability, while a high-fiber diet/short-chain fatty acids (SCFAs) can promote the development of Foxp3 Tregs and improvement in intestinal barrier function, which subsequently suppress CNS autoimmunity in the animal model of MS (experimental autoimmune encephalomyelitis or EAE). This review will address the role of gut immunity and its modulation by diet and DMTs via gut microbiota during MS pathophysiology.


Assuntos
Encefalomielite Autoimune Experimental , Microbioma Gastrointestinal , Esclerose Múltipla , Animais , Esclerose Múltipla/terapia , Microbioma Gastrointestinal/fisiologia , Sistema Nervoso Central , Dieta , Disbiose
6.
Crit Rev Food Sci Nutr ; 62(6): 1427-1452, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33198506

RESUMO

The oral antibiotic therapies administered widely to people and animals can cause gut dysbiosis and barrier disruption inevitably. Increasing attention has been directed toward antibiotic-induced gut dysbiosis, which involves a loss of diversity, changes in the abundances of certain taxa and consequent effects on their metabolic capacity, and the spread of antibiotic-resistant bacterial strains. Treatment with beta-lactam, glycopeptide, and macrolide antibiotics is associated with the depletion of beneficial commensal bacteria in the genera Bifidobacterium and Lactobacillus. The gut microbiota is a reservoir for antibiotic resistance genes, the prevalence of which increases sharply after antibiotic ingestion. The intestinal barrier, which comprises secretory, physical, and immunological barriers, is also a target of antibiotics. Antibiotic induced changes in the gut microbiota composition could induce weakening of the gut barrier through changes in mucin, cytokine, and antimicrobial peptide production by intestinal epithelial cells. Reports have indicated that dietary interventions involving prebiotics, probiotics, omega-3 fatty acids, and butyrate supplementation, as well as fecal microbiota transplantation, can alleviate antibiotic-induced gut dysbiosis and barrier injuries. This review summarizes the characteristics of antibiotic-associated gut dysbiosis and barrier disruption, as well as the strategies for alleviating this condition. This information is intended to provide a foundation for the exploration of safer, more efficient, and affordable strategies to prevent or relieve antibiotic-induced gut injuries.


Assuntos
Microbioma Gastrointestinal , Probióticos , Animais , Antibacterianos/toxicidade , Disbiose/induzido quimicamente , Disbiose/prevenção & controle , Humanos , Lactobacillus
7.
Appl Microbiol Biotechnol ; 106(21): 6899-6913, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36190540

RESUMO

Constipation is very pervasive all over the world. It is a common multifactorial gastrointestinal disease, and its etiology and pathomechanism are not completely clear. Now, increasing evidence shows that intestinal flora is closely related to constipation. Intestinal flora is the largest microbiota in the human body and has powerful metabolic functions. Intestinal flora can produce a variety of metabolites, such as bile acids, short-chain fatty acids, tryptophan metabolites, and methane, which have important effects on intestinal motility and secretion. The host can also monitor the intestinal flora and regulate gut dysbacteriosis in constipation. To explore the relationship between intestinal flora and host, the combination of multiomics technology has become the powerful and effective method. Furthermore, the homeostasis restoration of intestinal flora also provides a new strategy for the treatment of constipation. This review aims to explore the interaction between intestinal flora and host in constipation, which contributes to disclose the pathogenesis of constipation and the development of novel drugs for the treatment of constipation from the perspective of intestinal flora. KEY POINTS: • This review highlights the regulation of gut microbiota on the intestinal motility and secretion of host. • The current review gives an insight into the role of the host on the recognition and regulation of intestinal ecology under constipation. • The article also introduces some novel methods of current gut microbiota research and gut microbiota-based constipation therapies.


Assuntos
Microbioma Gastrointestinal , Humanos , Triptofano , Constipação Intestinal/metabolismo , Ácidos Graxos Voláteis , Ácidos e Sais Biliares/farmacologia , Metano
8.
J Allergy Clin Immunol ; 147(1): 335-348.e11, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32407834

RESUMO

BACKGROUND: The cross-talk between the host and its microbiota plays a key role in the promotion of health. The production of metabolites such as polyamines by intestinal-resident bacteria is part of this symbiosis shaping host immunity. The polyamines putrescine, spermine, and spermidine are abundant within the gastrointestinal tract and might substantially contribute to gut immunity. OBJECTIVE: We aimed to characterize the polyamine spermidine as a modulator of T-cell differentiation and function. METHODS: Naive T cells were isolated from wild-type mice or cord blood from healthy donors and submitted to polarizing cytokines, with and without spermidine treatment, to evaluate CD4+ T-cell differentiation in vitro. Moreover, mice were subjected to oral supplementation of spermidine, or its precursor l-arginine, to assess the frequency and total numbers of regulatory T (Treg) cells in vivo. RESULTS: Spermidine modulates CD4+ T-cell differentiation in vitro, preferentially committing naive T cells to a regulatory phenotype. After spermidine treatment, activated T cells lacking the autophagy gene Atg5 fail to upregulate Foxp3 to the same extent as wild-type cells. These results indicate that spermidine's polarizing effect requires an intact autophagic machinery. Furthermore, dietary supplementation with spermidine promotes homeostatic differentiation of Treg cells within the gut and reduces pathology in a model of T-cell transfer-induced colitis. CONCLUSION: Altogether, our results highlight the beneficial effects of spermidine, or l-arginine, on gut immunity by promoting Treg cell development.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Colite/imunologia , Imunidade nas Mucosas/efeitos dos fármacos , Espermidina/farmacologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout
9.
Int J Mol Sci ; 23(22)2022 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-36430509

RESUMO

Ulcerative colitis (UC) is a complex immune-mediated inflammatory disease. In recent years, the incidence of UC has increased rapidly, however, its exact etiology and mechanism are still unclear. Based on the definite anti-inflammatory and antibacterial activities of Sanguisorba officinalis L., we studied its monomer, methyl gallate (MG). In this study, we employed flow cytometry and detected nitric oxide production, finding MG regulated macrophage polarization and inhibited the expression of proinflammatory cytokines in vitro. MG also exhibited anti-inflammatory activity accompanying with ameliorating body weight loss, improving colon length and histological damage in dextran sulfate sodium-induced UC mice. Meanwhile, transcription sequencing and 16S rRNA sequencing analyzed the key signaling pathways and changes in the gut microbiota of MG for UC treatment, proving that MG could alleviate inflammation by regulating the TLR4/NF-κB pathway in vivo and in vitro. Additionally, MG altered the diversity and composition of the gut microbiota and changed the abundance of metabolic products. In conclusion, our results are the first to demonstrate that MG has obvious therapeutic effects against acute UC, which is related to macrophage polarization, improved intestinal flora dysbiosis and inhibition of TLR4/NF-κB signaling pathway, and MG may be a promising therapeutic agent for UC treatment.


Assuntos
Colite Ulcerativa , Microbioma Gastrointestinal , Camundongos , Animais , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/tratamento farmacológico , NF-kappa B , Receptor 4 Toll-Like , RNA Ribossômico 16S
10.
J Appl Microbiol ; 130(4): 1035-1061, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32869386

RESUMO

The human gastrointestinal (GI) tract has been bestowed with the most difficult task of protecting the underlying biological compartments from the resident commensal flora and the potential pathogens in transit through the GI tract. It has a unique environment in which several defence tactics are at play while maintaining homeostasis and health. The GI tract shows myriad number of environmental extremes, which includes pH variations, anaerobic conditions, nutrient limitations, elevated osmolarity etc., which puts a check to colonization and growth of nonfriendly microbial strains. The GI tract acts as a highly selective barrier/platform for ingested food and is the primary playground for balance between the resident and uninvited organisms. This review focuses on antimicrobial defense mechanisms of different sections of human GI tract. In addition, the protective mechanisms used by microbes to combat the human GI defence systems are also discussed. The ability to survive this innate defence mechanism determines the capability of probiotic or pathogen strains to confer health benefits or induce clinical events respectively.


Assuntos
Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Antibacterianos/análise , Antibacterianos/imunologia , Microbioma Gastrointestinal , Trato Gastrointestinal/química , Interações entre Hospedeiro e Microrganismos , Humanos , Imunidade Inata , Probióticos , Estresse Fisiológico
11.
J Appl Microbiol ; 130(5): 1425-1441, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33022786

RESUMO

Human gut microbiome is a diversified, resilient, immuno-stabilized, metabolically active and physiologically essential component of the human body. Scientific explorations have been made to seek in-depth information about human gut microbiome establishment, microbiome functioning, microbiome succession, factors influencing microbial community dynamics and the role of gut microbiome in health and diseases. Extensive investigations have proposed the microbiome therapeutics as a futuristic medicine for various physiological and metabolic disorders. A comprehensive outlook of microbial colonization, host-microbe interactions, microbial adaptation, commensal selection and immuno-survivability is still required to catalogue the essential genetic and physiological features for the commensal engagement. Evolution of a structured human gut microbiome relies on the microbial flexibility towards genetic, immunological and physiological adaptation in the human gut. Key features for commensalism could be utilized in developing tailor-made microbiome-based therapy to overcome various physiological and metabolic disorders. This review describes the key genetics and physiological traits required for host-microbe interaction and successful commensalism to institute a human gut microbiome.


Assuntos
Fenômenos Fisiológicos Bacterianos , Disbiose/terapia , Microbioma Gastrointestinal/fisiologia , Simbiose , Adaptação Fisiológica , Bactérias/genética , Bactérias/imunologia , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/imunologia , Interações entre Hospedeiro e Microrganismos , Humanos , Tolerância Imunológica , Redes e Vias Metabólicas , Estresse Fisiológico
12.
Biosci Biotechnol Biochem ; 85(6): 1536-1545, 2021 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-33885732

RESUMO

We analyzed the mechanisms underlying enhanced IgA production in the cells of Peyer's patch cells via membrane vesicles derived from Lactobacillus sakei subsp. sakei NBRC 15893. Depletion of CD11c+ cells from Peyer's patch cells suppressed the enhanced IgA production mediated by membrane vesicles. Meanwhile, the stimulation of bone-marrow-derived dendritic cells with membrane vesicles increased gene expression of inducible nitric oxide synthase, retinaldehyde dehydrogenase 2, and several inflammatory cytokines. The production of nitric oxide and interleukin (IL)-6 by membrane vesicle stimulation was induced via Toll-like receptor 2 on bone marrow-derived dendritic cells. Inhibition of inducible nitric oxide synthase and retinaldehyde dehydrogenase 2, as well as the neutralization of IL-6 in Peyer's patch cells, suppressed the enhanced IgA production by membrane vesicle stimulation. Hence, nitric oxide, retinoic acid, and IL-6 induced by membrane vesicles play crucial roles in the enhanced IgA production elicited by membrane vesicles in Peyer's patch cells.


Assuntos
Membrana Celular/metabolismo , Imunoglobulina A/biossíntese , Latilactobacillus sakei/citologia , Nódulos Linfáticos Agregados/metabolismo , Nódulos Linfáticos Agregados/citologia
13.
Int J Mol Sci ; 22(17)2021 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-34502262

RESUMO

Intestinal microfold cells (M cells) are a dynamic lineage of epithelial cells that initiate mucosal immunity in the intestine. They are responsible for the uptake and transcytosis of microorganisms, pathogens, and other antigens in the gastrointestinal tract. A mature M cell expresses a receptor Gp2 which binds to pathogens and aids in the uptake. Due to the rarity of these cells in the intestine, their development and differentiation remain yet to be fully understood. We recently demonstrated that polycomb repressive complex 2 (PRC2) is an epigenetic regulator of M cell development, and 12 novel transcription factors including Atoh8 were revealed to be regulated by the PRC2. Here, we show that Atoh8 acts as a regulator of M cell differentiation; the absence of Atoh8 led to a significant increase in the number of Gp2+ mature M cells and other M cell-associated markers such as Spi-B and Sox8. In vitro organoid analysis of RankL treated organoid showed an increase of mature marker GP2 expression and other M cell-associated markers. Atoh8 null mice showed an increase in transcytosis capacity of luminal antigens. An increase in M cell population has been previously reported to be detrimental to mucosal immunity because some pathogens like orally acquired prions have been able to exploit the transcytosis capacity of M cells to infect the host; mice with an increased population of M cells are also susceptible to Salmonella infections. Our study here demonstrates that PRC2 regulated Atoh8 is one of the factors that regulate the population density of intestinal M cell in the Peyer's patch.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/genética , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Animais , Linfócitos B/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/imunologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Imunidade nas Mucosas/genética , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Camundongos , Camundongos Knockout , Nódulos Linfáticos Agregados/efeitos dos fármacos , Nódulos Linfáticos Agregados/metabolismo , Cultura Primária de Células , Ligante RANK/farmacologia , Receptor Ativador de Fator Nuclear kappa-B/farmacologia , Linfócitos T/metabolismo , Transcitose/genética
14.
Int J Mol Sci ; 22(4)2021 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-33672304

RESUMO

Communication between cells is crucial to preserve body homeostasis and health. Tightly controlled intercellular dialog is particularly relevant in the gut, where cells of the intestinal mucosa are constantly exposed to millions of microbes that have great impact on intestinal homeostasis by controlling barrier and immune functions. Recent knowledge involves extracellular vesicles (EVs) as mediators of such communication by transferring messenger bioactive molecules including proteins, lipids, and miRNAs between cells and tissues. The specific functions of EVs principally depend on the internal cargo, which upon delivery to target cells trigger signal events that modulate cellular functions. The vesicular cargo is greatly influenced by genetic, pathological, and environmental factors. This finding provides the basis for investigating potential clinical applications of EVs as therapeutic targets or diagnostic biomarkers. Here, we review current knowledge on the biogenesis and cargo composition of EVs in general terms. We then focus the attention to EVs released by cells of the intestinal mucosa and their impact on intestinal homeostasis in health and disease. We specifically highlight their role on epithelial barrier integrity, wound healing of epithelial cells, immunity, and microbiota shaping. Microbiota-derived EVs are not reviewed here.


Assuntos
Vesículas Extracelulares/metabolismo , Microbioma Gastrointestinal/fisiologia , Mucosa Intestinal/fisiologia , Intestinos/citologia , MicroRNAs/imunologia , Animais , Comunicação Celular , Proliferação de Células , Vesículas Extracelulares/química , Vesículas Extracelulares/classificação , Vesículas Extracelulares/genética , Humanos , Células-Tronco Mesenquimais/citologia
15.
Biogerontology ; 21(5): 609-618, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32277312

RESUMO

The chemokines CCL25 and CCL28, which promote immune cell migration, are primarily expressed in the small and large intestines and play critical roles in sustaining gut immunity. In particular, these chemokines are closely related to intestinal IgA secretion. However, there is no research regarding the effects of aging on CCL25 and CCL28 expression and function. Therefore, in the present study, we investigated the effects of aging on production of CCL25 and CCL28, and on gut immunity, especially IgA secretion, using young and aged female mice. By aging, the levels of small intestinal mRNA and protein of CCL25 lowered, while these levels of CCL28 in colon became higher. Moreover, the number of IgA-antibody secreting cells (IgA-ASCs) and total IgA concentration decreased in the small intestine due to the age-associated reduction of CCL25. In contrast, colonic IgA production was increased due to up-regulation of CCL28, while the number of colonic IgA-ASCs was unchanged with aging. These results clearly demonstrate that aging-associated decrease in small intestinal CCL25 production and increase in colonic CCL28 production c be involved in aging-associated deterioration of gut immunity.


Assuntos
Envelhecimento/imunologia , Quimiocinas , Imunidade nas Mucosas , Imunoglobulina A , Animais , Movimento Celular , Quimiocinas/imunologia , Quimiocinas CC , Colo/imunologia , Feminino , Imunoglobulina A/imunologia , Camundongos , RNA Mensageiro
16.
Neuroimmunomodulation ; 26(6): 265-275, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31715599

RESUMO

Mental health is becoming a public health priority worldwide. Anorexia nervosa and autism spectrum disorders are 2 important types of childhood disorders with a bad prognosis. They share cognitive impairments and, in both cases, the microbiota appears to be a crucial factor. Alteration of the microbiota-gut-brain axis is an appealing hypothesis to define new pathophysiological mechanisms. Mucosal immunity plays a key role between the microbiota and the brain. The mucosal immune system receives and integrates messages from the intestinal microenvironment and the microbiota and then transmits the information to the nervous system. Abnormalities in this sensorial system may be involved in the natural history of mental diseases and might play a role in their maintenance. This review aims to highlight data about the relationship between intestinal mucosal immunity and these disorders. We show that shared cognitive impairments could be found in these 2 disorders, which both present dysbiosis. This literature review provides details on the immune status of anorexic and autistic patients, with a focus on intestinal mucosal factors. Finally, we suggest future research hypotheses that seem important for understanding the implication of the gut-brain-axis in psychiatric diseases.


Assuntos
Anorexia Nervosa/imunologia , Transtorno do Espectro Autista/imunologia , Microbioma Gastrointestinal/imunologia , Imunidade nas Mucosas/imunologia , Mucosa Intestinal/imunologia , Neuroimunomodulação/imunologia , Animais , Anorexia Nervosa/microbiologia , Transtorno do Espectro Autista/microbiologia , Humanos , Mucosa Intestinal/microbiologia
17.
Fish Shellfish Immunol ; 89: 368-377, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30965086

RESUMO

Recent studies in mammalian models revealed compelling evidence that along with the intrinsic characteristics of diets, the time of their delivery could have a profound impact on their benefits. In this study, we explored a time-dependent modulation of the gut mucosal barrier by delivering diets enriched with the green microalga (Lobosphaera incisa) either in a time-restricted regime or randomly to zebrafish (Danio rerio). The basal diet was enriched with microalgal biomass through two inclusion levels (i.e., 10% and 15% w/w), and the feeding trial lasted for six weeks. The control group was fed with the basal diet. After collection of tissue samples at week 6, the remaining fish were challenged by intraperitoneal injection of Streptococcus inaie. A histological analysis of the gut structure revealed that the fish that received the microalgae randomly exhibited shorter villi length. Genes coding for immunity were modulated in the gut by dietary treatments. Notably, the transcript levels of lysozyme, ß-defensin and hepcidin were significantly higher in the group subjected to the time-restricted feeding regime. Dietary microalgae affected the fatty acid content in the gut, particularly the level of arachidonic acid (ARA), and the time-restricted feeding influenced its accumulation. Groups that received diets enriched with 15% microalgae, regardless of the feeding strategy, displayed a significantly higher resistance to S. inaie 16 days post-infection, though differences between the delivery strategies were pronounced during the early stage of infection. In conclusion, the dietary inclusion of L. incisa modulated some of the features of the gut mucosal barrier of zebrafish, and the time of delivery appeared to have a considerable influence on immunomodulatory functions.


Assuntos
Ração Animal/análise , Criação de Animais Domésticos/métodos , Clorófitas , Mucosa Intestinal/imunologia , Peixe-Zebra/imunologia , Animais , Clorófitas/química , Dieta/veterinária , Microalgas/química , Fatores de Tempo
18.
Digestion ; 99(4): 267-274, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30293081

RESUMO

BACKGROUND: Gastrointestinal (GI) diseases are known to be largely influenced by one's lifestyle and dietary uptake. A high-salt diet (HSD) is well recognized as a risk factor for cardiovascular complications, hypertension, and metabolic syndromes. However, the relationship between an HSD and the GI system, which is the compartment that comes in direct contact with exogenous stimulants, has not been fully explored. AIMS: We seek to better understand the complexity of the pathogenic effects of an HSD in the context of GI disorders. METHODS: By searching the PubMed and Web of science, the review of literature was performed using keywords: high-salt and GI, high-salt and immunity, salt and microbiota, salt and hormone. RESULTS: In this review, we concluded that high-salt intake potentially perturbs the local immune homeostasis, alters the gut microbiota composition and function, and affects the endocrine hormone profiling in the GI system. CONCLUSION: HSD might get involved in GI diseases through the reshaping of gastroenterological milieu, which could help to better understand the complexity of the pathogenic effects of an HSD in the context of GI disorders.


Assuntos
Comportamento Alimentar/fisiologia , Gastroenteropatias/epidemiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Sódio na Dieta/efeitos adversos , Gastroenteropatias/etiologia , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiologia , Humanos , Incidência , Fatores de Risco
19.
Biochem Biophys Res Commun ; 495(1): 395-400, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29108998

RESUMO

In this study, fruit fly of the genus Drosophila is utilized as a suitable model animal to investigate the molecular mechanisms of innate immunity. To combat orally transmitted pathogenic Gram-negative bacteria, the Drosophila gut is armed with the peritrophic matrix, which is a physical barrier composed of chitin and glycoproteins: the Duox system that produces reactive oxygen species (ROS), which in turn sterilize infected microbes, and the IMD pathway that regulates the expression of antimicrobial peptides (AMPs), which in turn control ROS-resistant pathogens. However, little is known about the defense mechanisms against Gram-positive bacteria in the fly gut. Here, we show that the peritrophic matrix protects Drosophila against Gram-positive bacteria S. aureus. We also define the few roles of ROS in response to the infection and show that the IMD pathway is required for the clearance of ingested microbes, possibly independently from AMP expression. These findings provide a new aspect of the gut defense system of Drosophila, and helps to elucidate the processes of gut-microbe symbiosis and pathogenesis.


Assuntos
Drosophila melanogaster/imunologia , Drosophila melanogaster/microbiologia , Interações Hospedeiro-Patógeno , Transdução de Sinais , Staphylococcus aureus/imunologia , Animais , Peptídeos Catiônicos Antimicrobianos/imunologia , Proteínas de Drosophila/imunologia , Feminino , Imunidade Inata , Masculino , Espécies Reativas de Oxigênio/imunologia
20.
Brain Behav Immun ; 68: 123-131, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29032226

RESUMO

Secretory Immunoglobulin A (sIgA) plays a critical role to infant gut mucosal immunity. Delayed IgA production is associated with greater risk of allergic disease. Murine models of stressful events during pregnancy and infancy show alterations in gut immunity and microbial composition in offspring, but little is known about the stress-microbiome-immunity pathways in humans. We investigated differences in infant fecal sIgA concentrations according to the presence of maternal depressive symptoms during and after pregnancy. A subsample of 403 term infants from the Canadian Healthy Infant Longitudinal Development (CHILD) cohort were studied. Their mothers completed the Center of Epidemiologic Studies Depression Scale when enrolled prenatally and again postpartum. Quantified by Immundiagnostik sIgA ELISA kit, sIgA from infant stool was compared across maternal depressive symptom categories using Mann-Whitney U-tests and logistic regression models that controlled for various covariates. Twelve percent of women reported clinically significant depressive symptoms only prenatally, 8.7% had only postpartum symptoms and 9.2% had symptoms both pre and postnatally. Infants born to mothers with pre and postnatal symptoms had significantly lower median sIgA concentrations than those in the reference group (4.4 mg/g feces vs. 6.3 mg/g feces; p = 0.033). The odds for sIgA concentrations in the lowest quartile was threefold higher (95% CI: 1.25-7.55) when mothers had pre and postnatal symptoms, after controlling for breastfeeding status, infant age, antibiotics exposure and other covariates. Postnatal symptoms were not associated with fecal sIgA, independently of breastfeeding status. Infants born to mothers with depressive symptoms appear to have lower fecal sIgA concentrations, predisposing them to higher risk for allergic disease.


Assuntos
Depressão Pós-Parto/metabolismo , Depressão/metabolismo , Imunoglobulina A Secretora/metabolismo , Adulto , Canadá , Estudos de Coortes , Fezes , Feminino , Microbioma Gastrointestinal , Humanos , Imunidade nas Mucosas , Imunoglobulina A/análise , Imunoglobulina A/metabolismo , Imunoglobulina A Secretora/análise , Lactente , Recém-Nascido , Masculino , Mães , Período Pós-Parto , Gravidez , Escalas de Graduação Psiquiátrica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa