Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 244
Filtrar
1.
J Extracell Vesicles ; 11(1): e12172, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34981901

RESUMO

Intestinal commensal bacteria contribute to maintaining gut homeostasis. Disruptions to the commensal flora are linked to the development and persistence of disease. The importance of these organisms is further demonstrated by the widespread ability of enteric viruses to exploit commensal bacteria to enhance viral infection. These viruses interact directly with commensal bacteria, and while the impact of this interaction on viral infection is well described for several viruses, the impact on the commensal bacteria has yet to be explored. In this article, we demonstrate, for the first time, that enteric viruses alter the gene expression and phenotype of individual commensal bacteria. Human and murine norovirus interaction with bacteria resulted in genome-wide differential gene expression and marked changes in the surface architecture of the bacterial cells. Furthermore, the interaction of the virus with bacteria led to increased production of smaller outer membrane vesicles (OMVs). Enhanced production of smaller vesicles was also observed when noroviruses were incubated with other commensal bacteria, indicating a potentially broad impact of norovirus interaction. The vesicle production observed in the in vivo model followed a similar trend where an increased quantity of smaller bacterial vesicles was observed in stool collected from virus-infected mice compared to mock-infected mice. Furthermore, changes in vesicle size were linked to changes in protein content and abundance, indicating that viral binding induced a shift in the mechanism of the OMV biogenesis. Collectively, these data demonstrate that enteric viruses induce specific changes in bacterial gene expression, leading to changes in bacterial extracellular vesicle production that can potentially impact host responses to infection.


Assuntos
Membrana Externa Bacteriana/metabolismo , Vesículas Extracelulares/metabolismo , Gastroenterite/microbiologia , Microbioma Gastrointestinal , Norovirus/fisiologia , Animais , Membrana Externa Bacteriana/ultraestrutura , Enterobacter cloacae/genética , Enterobacter cloacae/metabolismo , Gastroenterite/metabolismo , Gastroenterite/virologia , Humanos , Camundongos , Interações Microbianas
2.
Int Arch Allergy Immunol ; 183(1): 80-92, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34515121

RESUMO

INTRODUCTION: The increase in high-fat diet (HFD)-induced obesity and food allergy leads to an assumption that the 2 are related. This study aims to (1) systematic verification of HFD-induced obesity aggravates food allergy and (2) explore the correlation and molecular mechanisms of HFD-induced obesity promotes food allergy. METHODS: Female BALB/c mice are divided into the control group (control), the ovalbumin (OVA)-sensitized group (OVA), the HFD-induced obesity group (HFD), and HFD-induced allergic obesity group (HFD + OVA). RESULTS: In vivo data showed that HFD feed enhance clinical symptoms and intestinal mucosa villi shed on allergic mice. Moreover, we found that HFD and OVA irritation enhanced levels of mast cell degranulation and Th2 humoral response. Additionally, Western blot analysis showed the potentiation of peroxisome proliferator-activated receptor γ (PPAR γ) remarkably reduced on intestinal in HFD and OVA group, thereby inhibiting the expression of nuclear factor kappa B (NF-κB)/PPAR γ signal the phosphorylation of NF-κB P65. CONCLUSIONS: Overall, our results suggest that HFD-induced obesity is a potential risk factor for food allergy, which related to intestinal barrier destruction and inflammation through the PPAR γ/NF-κB signaling pathway.


Assuntos
Hipersensibilidade Alimentar/etiologia , Hipersensibilidade Alimentar/metabolismo , Gastroenterite/etiologia , Gastroenterite/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Obesidade/complicações , Animais , Biomarcadores , Citocinas/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Hipersensibilidade Alimentar/patologia , Gastroenterite/patologia , Imuno-Histoquímica , Mucosa Intestinal/patologia , Camundongos , NF-kappa B/metabolismo , Obesidade/etiologia , PPAR gama , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
3.
Sci Rep ; 11(1): 11788, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34083551

RESUMO

This study investigated the immune mechanisms whereby administration of Bacteroides uniformis CECT 7771 reduces metabolic dysfunction in obesity. C57BL/6 adult male mice were fed a standard diet or a Western diet high in fat and fructose, supplemented or not with B. uniformis CECT 7771 for 14 weeks. B. uniformis CECT 7771 reduced body weight gain, plasma cholesterol, triglyceride, glucose, and leptin levels; and improved oral glucose tolerance in obese mice. Moreover, B. uniformis CECT 7771 modulated the gut microbiota and immune alterations associated with obesity, increasing Tregs and reducing B cells, total macrophages and the M1/M2 ratio in both the gut and epididymal adipose tissue (EAT) of obese mice. B. uniformis CECT 7771 also increased the concentration of the anti-inflammatory cytokine IL-10 in the gut, EAT and peripheral blood, and protective cytokines TSLP and IL-33, involved in Treg induction and type 2 innate lymphoid cells activation, in the EAT. It also restored the obesity-reduced TLR5 expression in the ileum and EAT. The findings indicate that the administration of a human intestinal bacterium with immunoregulatory properties on the intestinal mucosa helps reverse the immuno-metabolic dysfunction caused by a Western diet acting over the gut-adipose tissue axis.


Assuntos
Infecções por Bacteroides/metabolismo , Infecções por Bacteroides/microbiologia , Bacteroides/fisiologia , Gastroenterite/metabolismo , Gastroenterite/microbiologia , Transdução de Sinais , Receptor 5 Toll-Like/metabolismo , Imunidade Adaptativa , Tecido Adiposo/metabolismo , Animais , Infecções por Bacteroides/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Metabolismo Energético , Gastroenterite/patologia , Microbioma Gastrointestinal , Imunidade Inata , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Obesos , Fenótipo
4.
Front Immunol ; 12: 668654, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054838

RESUMO

Chronic low-grade inflammation is a hallmark of obesity and associated with cardiovascular complications. However, it remains unclear where this inflammation starts. As the gut is constantly exposed to food, gut microbiota, and metabolites, we hypothesized that mucosal immunity triggers an innate inflammatory response in obesity. We characterized five distinct macrophage subpopulations (P1-P5) along the gastrointestinal tract and blood monocyte subpopulations (classical, non-classical, intermediate), which replenish intestinal macrophages, in non-obese (BMI<27kg/m2) and obese individuals (BMI>32kg/m2). To elucidate factors that potentially trigger gut inflammation, we correlated these subpopulations with cardiovascular risk factors and lifestyle behaviors. In obese individuals, we found higher pro-inflammatory macrophages in the stomach, duodenum, and colon. Intermediate blood monocytes were also increased in obesity, suggesting enhanced recruitment to the gut. We identified unhealthy lifestyle habits as potential triggers of gut and systemic inflammation (i.e., low vegetable intake, high processed meat consumption, sedentary lifestyle). Cardiovascular risk factors other than body weight did not affect the innate immune response. Thus, obesity in humans is characterized by gut inflammation as shown by accumulation of pro-inflammatory intestinal macrophages, potentially via recruited blood monocytes. Understanding gut innate immunity in human obesity might open up new targets for immune-modulatory treatments in metabolic disease.


Assuntos
Gastroenterite/imunologia , Imunidade Inata , Imunidade nas Mucosas , Intestinos/imunologia , Macrófagos/imunologia , Obesidade/imunologia , Índice de Massa Corporal , Estudos de Casos e Controles , Dieta/efeitos adversos , Feminino , Gastroenterite/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/metabolismo , Fenótipo , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Comportamento Sedentário
5.
Front Immunol ; 12: 578386, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717063

RESUMO

Hosting millions of microorganisms, the digestive tract is the primary and most important part of bacterial colonization. On one side, in cases of opportunistic invasion, the abundant bacterial population inside intestinal tissues may face potential health problems such as inflammation and infections. Therefore, the immune system has evolved to sustain the host-microbiota symbiotic relationship. On the other hand, to maintain host immune homeostasis, the intestinal microflora often exerts an immunoregulatory function that cannot be ignored. A field of great interest is the association of either microbiota or probiotics with the immune system concerning clinical uses. This microbial community regulates some of the host's metabolic and physiological functions and drives early-life immune system maturation, contributing to their homeostasis throughout life. Changes in gut microbiota can occur through modification in function, composition (dysbiosis), or microbiota-host interplays. Studies on animals and humans show that probiotics can have a pivotal effect on the modulation of immune and inflammatory mechanisms; however, the precise mechanisms have not yet been well defined. Diet, age, BMI (body mass index), medications, and stress may confound the benefits of probiotic intake. In addition to host gut functions (permeability and physiology), all these agents have profound implications for the gut microbiome composition. The use of probiotics could improve the gut microbial population, increase mucus-secretion, and prevent the destruction of tight junction proteins by decreasing the number of lipopolysaccharides (LPSs). When LPS binds endothelial cells to toll-like receptors (TLR 2, 4), dendritic cells and macrophage cells are activated, and inflammatory markers are increased. Furthermore, a decrease in gut dysbiosis and intestinal leakage after probiotic therapy may minimize the development of inflammatory biomarkers and blunt unnecessary activation of the immune system. In turn, probiotics improve the differentiation of T-cells against Th2 and development of Th2 cytokines such as IL-4 and IL-10. The present narrative review explores the interactions between gut microflora/probiotics and the immune system starting from the general perspective of a biological plausibility to get to the in vitro and in vivo demonstrations of a probiotic-based approach up to the possible uses for novel therapeutic strategies.


Assuntos
Anti-Inflamatórios/farmacologia , Gastroenterite/etiologia , Imunomodulação/efeitos dos fármacos , Probióticos/administração & dosagem , Animais , Anti-Inflamatórios/uso terapêutico , Dieta , Suscetibilidade a Doenças , Disbiose , Gastroenterite/tratamento farmacológico , Gastroenterite/metabolismo , Gastroenterite/patologia , Microbioma Gastrointestinal/imunologia , Humanos , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Estilo de Vida
6.
Front Immunol ; 12: 619262, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717115

RESUMO

Background: Oral-gut inflammation has an impact on overall health, placing subjects at risk to acquire chronic conditions and infections. Due to neuromotor disturbances, and medication intake, cerebral palsy (CP) subjects present intestinal constipation, impacting their quality of life (QOL). We aimed to investigate how oral inflammatory levels predicted gut phenotypes and response to therapy. Methods: A total of 93 subjects aging from 5 to 17 years were included in the study, and assigned into one of the 4 groups: CP with constipation (G1, n = 30), CP without constipation (G2, n = 33), and controls without CP with constipation (G3, n = 07) and without CP and without constipation (G4, n = 23). In addition to characterizing subjects' clinical demographics, medication intake, disease severity levels, salivary cytokine levels [TNF-α, interleukin (IL)-1ß, IL-6, IL-8, IL-10], and Caregiver Priorities and Child Health Index of Life with Disabilities (CPCHILD). Statistical significance was evaluated by Shapiro-Wilks, Student's T-Test, ANOVA, and ANCOVA analysis. Results: Salivary proinflammatory cytokines were highly correlated with the severe form of gut constipation in G1 (P < 0.001), and out of all cytokines IL-1ß levels demonstrated highest correlation with all gut constipation (P < 0.05). A significant relationship was found between the type of medication, in which subjects taking Gamma-Aminobutyric Acid (GABA) and GABA+ (GABA in association with other medication) were more likely to be constipated than the other groups (P < 0.01). Cleary salivary inflammatory levels and gut constipation were correlated, and impacted QOL of CP subjects. G1 presented a lower QOL mean score of CPCHILD (49.0 ± 13.1) compared to G2 (71.5 ± 16.7), when compared to G3 (88.9 ± 7.5), and G4 (95.5 ± 5.0) (P < 0.01). We accounted for gingival bleeding as a cofounder of oral inflammation, and here were no differences among groups regarding gender (P = 0.332) and age (P = 0.292). Conclusions: Collectively, the results suggest that saliva inflammatory levels were linked to gut constipation, and that the clinical impact of medications that controlled gut was reliably monitored via oral cytokine levels, providing reliable and non-invasive information in precision diagnostics.


Assuntos
Paralisia Cerebral/complicações , Paralisia Cerebral/epidemiologia , Gastroenterite/complicações , Gastroenterite/epidemiologia , Estomatite/complicações , Estomatite/epidemiologia , Adolescente , Biomarcadores , Paralisia Cerebral/metabolismo , Criança , Pré-Escolar , Estudos Transversais , Citocinas/metabolismo , Feminino , Gastroenterite/diagnóstico , Gastroenterite/metabolismo , Humanos , Mediadores da Inflamação , Masculino , Fenótipo , Vigilância da População , Qualidade de Vida , Saliva/metabolismo , Estomatite/diagnóstico , Estomatite/metabolismo , Avaliação de Sintomas
7.
Int J Biol Macromol ; 174: 289-299, 2021 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-33524482

RESUMO

Capsaicin (CAP) is the main pungent component in capsicum fruits. Eating too much CAP leads to gastrointestinal injury. Previously, Qingke ß-glucan combined with ß-glucan-utilizing Lactobacillus plantarum S58 (LP.S58) ameliorated high fat-diet-induced obesity, but their effects on CAP-induced gastrointestinal injury have not been investigated. Our results showed that Qingke ß-glucan reduced the CAP-induced gastrointestinal injury in Kunming mice. The serum levels of inflammatory cytokines and gastrointestinal hormones, and the localized inflammation and the expression of EGF, EGFR, VEGF, and ZO-1 in the gastrointestinal tissues in CAP-treated mice were partly restored by Qingke ß-glucan. The CAP-induced increase in the abundances of proinflammatory intestinal bacteria was also reduced by Qingke ß-glucan. More importantly, we found that these beneficial effects of Qingke ß-glucan were markedly enhanced by ß-glucan-utilizing LP.S58 supplementation. Our study indicated that Qingke ß-glucan coupled with ß-glucan-utilizing LP.S58 relieved CAP-induced gastrointestinal injury.


Assuntos
Capsaicina/efeitos adversos , Gastroenterite/terapia , Glucanos/administração & dosagem , Hordeum/química , Lactobacillus plantarum/fisiologia , Probióticos/administração & dosagem , Animais , Terapia Combinada , Citocinas/sangue , Modelos Animais de Doenças , Sinergismo Farmacológico , Receptores ErbB/genética , Receptores ErbB/metabolismo , Gastroenterite/induzido quimicamente , Gastroenterite/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Glucanos/metabolismo , Glucanos/farmacologia , Lactobacillus plantarum/metabolismo , Masculino , Camundongos , Extratos Vegetais/administração & dosagem , Extratos Vegetais/metabolismo , Extratos Vegetais/farmacologia , Probióticos/farmacologia , Resultado do Tratamento , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
8.
J Virol ; 95(3)2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33115870

RESUMO

Human noroviruses are the most common nonbacterial cause of gastroenteritis outbreaks, with new variants and genotypes frequently emerging. The origin of these new viruses is unknown; however, animals have been proposed as a potential source, as human noroviruses have been detected in animal species. Here, we investigated the potential of animals to serve as a reservoir of human noroviruses by testing norovirus attachment to formalin-fixed intestinal tissues of a range of potential reservoir animals. We set up a novel method to study norovirus binding using fluorescein isothiocyanate (FITC)-labeled virus-like particles (VLPs). In humans, noroviruses interact with histo-blood group antigens (HBGAs), carbohydrates that are expressed, among others, on the epithelial lining of the gastrointestinal tract. In animals, this interaction is not well understood. To test if virus binding depends on HBGAs, we characterized the HBGA phenotype in animal tissues by immunohistochemistry. With the exception of the black-headed gull and the straw-colored fruitbat, we observed the attachment of several human norovirus genotypes to the intestinal epithelium of all tested animal species. However, we did not find an association between the expression of a specific HBGA phenotype and virus-like particle (VLP) attachment. We show that selected human noroviruses can attach to small-intestinal tissues across species, supporting the hypothesis that human noroviruses can reside in an animal reservoir. However, whether this attachment can subsequently lead to infection needs to be further assessed.IMPORTANCE Noroviruses are a major cause of acute gastroenteritis in humans. New norovirus variants and recombinants (re)emerge regularly in the human population. From animal experiments and surveillance studies, it has become clear that at least seven animal models are susceptible to infection with human strains and that domesticated and wild animals shed human noroviruses in their feces. As virus attachment is an important first step for infection, we used a novel method utilizing FITC-labeled VLPs to test for norovirus attachment to intestinal tissues of potential animal hosts. We further characterized these tissues with regard to their HBGA expression, a well-studied norovirus susceptibility factor in humans. We found attachment of several human strains to a variety of animal species independent of their HBGA phenotype. This supports the hypothesis that human strains could reside in an animal reservoir.


Assuntos
Antígenos de Grupos Sanguíneos/metabolismo , Infecções por Caliciviridae/virologia , Modelos Animais de Doenças , Gastroenterite/virologia , Mucosa Intestinal/virologia , Norovirus/fisiologia , Ligação Viral , Sequência de Aminoácidos , Animais , Infecções por Caliciviridae/metabolismo , Infecções por Caliciviridae/patologia , Fezes/virologia , Gastroenterite/metabolismo , Gastroenterite/patologia , Humanos , Mucosa Intestinal/metabolismo , Homologia de Sequência
9.
Gastroenterology ; 160(5): 1647-1661, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33307034

RESUMO

BACKGROUND & AIMS: Gastrointestinal (GI) manifestations have been increasingly reported in patients with coronavirus disease 2019 (COVID-19). However, the roles of the GI tract in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are not fully understood. We investigated how the GI tract is involved in SARS-CoV-2 infection to elucidate the pathogenesis of COVID-19. METHODS: Our previously established nonhuman primate (NHP) model of COVID-19 was modified in this study to test our hypothesis. Rhesus monkeys were infected with an intragastric or intranasal challenge with SARS-CoV-2. Clinical signs were recorded after infection. Viral genomic RNA was quantified by quantitative reverse transcription polymerase chain reaction. Host responses to SARS-CoV-2 infection were evaluated by examining inflammatory cytokines, macrophages, histopathology, and mucin barrier integrity. RESULTS: Intranasal inoculation with SARS-CoV-2 led to infections and pathologic changes not only in respiratory tissues but also in digestive tissues. Expectedly, intragastric inoculation with SARS-CoV-2 resulted in the productive infection of digestive tissues and inflammation in both the lung and digestive tissues. Inflammatory cytokines were induced by both types of inoculation with SARS-CoV-2, consistent with the increased expression of CD68. Immunohistochemistry and Alcian blue/periodic acid-Schiff staining showed decreased Ki67, increased cleaved caspase 3, and decreased numbers of mucin-containing goblet cells, suggesting that the inflammation induced by these 2 types of inoculation with SARS-CoV-2 impaired the GI barrier and caused severe infections. CONCLUSIONS: Both intranasal and intragastric inoculation with SARS-CoV-2 caused pneumonia and GI dysfunction in our rhesus monkey model. Inflammatory cytokines are possible connections for the pathogenesis of SARS-CoV-2 between the respiratory and digestive systems.


Assuntos
COVID-19/transmissão , Gastroenterite/patologia , Trato Gastrointestinal/patologia , Pulmão/patologia , Animais , Brônquios/metabolismo , Brônquios/patologia , COVID-19/imunologia , COVID-19/metabolismo , COVID-19/patologia , Teste de Ácido Nucleico para COVID-19 , Caspase 3/metabolismo , Citocinas/imunologia , Modelos Animais de Doenças , Mucosa Gástrica , Gastroenterite/metabolismo , Gastroenterite/virologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/metabolismo , Células Caliciformes/patologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Antígeno Ki-67/metabolismo , Pulmão/diagnóstico por imagem , Pulmão/imunologia , Pulmão/metabolismo , Macaca mulatta , Mucosa Nasal , RNA Viral/isolamento & purificação , Distribuição Aleatória , Reto/metabolismo , Reto/patologia , SARS-CoV-2 , Traqueia/metabolismo , Traqueia/patologia
10.
Food Microbiol ; 92: 103594, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32950136

RESUMO

Human noroviruses (HuNoVs) are a main cause of acute gastroenteritis worldwide. They are frequently involved in foodborne and waterborne outbreaks. Environmental transmission of the virus depends on two main factors: the ability of viral particles to remain infectious and their adhesion capacity onto different surfaces. Until recently, adhesion of viral particles to food matrices was mainly investigated by considering non-specific interactions (e.g. electrostatic, hydrophobic) and there was only limited information about infectious HuNoVs because of the absence of a reliable in vitro HuNoV cultivation system. Many HuNoV strains have now been described as having specific binding interactions with human Histo-Blood Group Antigens (HBGAs) and non-HBGA ligands found in food and the environment. Relevant approaches to the in vitro replication of HuNoVs were also proposed recently. On the basis of the available literature data, this review discusses the opportunities to use this new knowledge to obtain a better understanding of HuNoV transmission to human populations and better evaluate the hazard posed by HuNoVs in foodstuffs and the environment.


Assuntos
Antígenos de Grupos Sanguíneos/metabolismo , Infecções por Caliciviridae/metabolismo , Gastroenterite/metabolismo , Norovirus/metabolismo , Animais , Antígenos de Grupos Sanguíneos/genética , Infecções por Caliciviridae/terapia , Infecções por Caliciviridae/transmissão , Infecções por Caliciviridae/virologia , Gastroenterite/genética , Gastroenterite/terapia , Gastroenterite/virologia , Humanos , Norovirus/genética , Norovirus/isolamento & purificação , Norovirus/fisiologia , Ligação Proteica , Proteínas Virais/genética , Proteínas Virais/metabolismo
11.
Acta Biochim Pol ; 67(3): 393-399, 2020 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-32945647

RESUMO

We compared fecal samples from responders and non-responders to administration of Lactobacillus reuteri DSM 17938. Data for this post hoc analysis were collected from an RCT assessing the efficacy of L. reuteri for the management of acute gastroenteritis. Responders were defined as subjects with diarrhea lasting no longer than 48 h. 44 children (17 responders and 27 non-responders) were analyzed. There were no differences in clinical characteristics and gut colonization between both groups. In the responder group, there were significantly lower levels of five metabolites before beginning of the intervention: lactate, choline, ethanol, creatine, and formate. The fecal calprotectin level did not differ between groups prior to the intervention, but its level was significantly lower after intervention in the responder group. Possibly, the responder group with a "metabolic niche", including lower level of metabolites, especially lactate, that are potential products of Lactobacillus genus, would determine the response to probiotic treatment. These findings need to be confirmed, but identification of some differences in the fecal metabolomics and the calprotectin level suggests that further studies are warranted.


Assuntos
Diarreia/dietoterapia , Suplementos Nutricionais/microbiologia , Fezes/química , Fezes/microbiologia , Gastroenterite/dietoterapia , Limosilactobacillus reuteri/metabolismo , Probióticos/uso terapêutico , Doença Aguda , Pré-Escolar , Diarreia/metabolismo , Método Duplo-Cego , Feminino , Gastroenterite/metabolismo , Humanos , Lactente , Complexo Antígeno L1 Leucocitário/análise , Complexo Antígeno L1 Leucocitário/metabolismo , Masculino , Metaboloma , Resultado do Tratamento
12.
J Virol ; 94(13)2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32321816

RESUMO

Human norovirus frequently causes outbreaks of acute gastroenteritis. Although discovered more than five decades ago, antiviral development has, until recently, been hampered by the lack of a reliable human norovirus cell culture system. Nevertheless, a lot of pathogenesis studies were accomplished using murine norovirus (MNV), which can be grown routinely in cell culture. In this study, we analyzed a sizeable library of nanobodies that were raised against the murine norovirus virion with the main purpose of developing nanobody-based inhibitors. We discovered two types of neutralizing nanobodies and analyzed the inhibition mechanisms using X-ray crystallography, cryo-electron microscopy (cryo-EM), and cell culture techniques. The first type bound on the top region of the protruding (P) domain. Interestingly, this nanobody binding region closely overlapped the MNV receptor-binding site and collectively shared numerous P domain-binding residues. In addition, we showed that these nanobodies competed with the soluble receptor, and this action blocked virion attachment to cultured cells. The second type bound at a dimeric interface on the lower side of the P dimer. We discovered that these nanobodies disrupted a structural change in the capsid associated with binding cofactors (i.e., metal cations/bile acid). Indeed, we found that capsids underwent major conformational changes following addition of Mg2+ or Ca2+ Ultimately, these nanobodies directly obstructed a structural modification reserved for a postreceptor attachment stage. Altogether, our new data show that nanobody-based inhibition could occur by blocking functional and structural capsid properties.IMPORTANCE This research discovered and analyzed two different types of MNV-neutralizing nanobodies. The top-binding nanobodies sterically inhibited the receptor-binding site, whereas the dimeric-binding nanobodies interfered with a structural modification associated with cofactor binding. Moreover, we found that the capsid contained a number of vulnerable regions that were essential for viral replication. In fact, the capsid appeared to be organized in a state of flux, which could be important for cofactor/receptor-binding functions. Blocking these capsid-binding events with nanobodies directly inhibited essential capsid functions. Moreover, a number of MNV-specific nanobody binding epitopes were comparable to human norovirus-specific nanobody inhibitors. Therefore, this additional structural and inhibition information could be further exploited in the development of human norovirus antivirals.


Assuntos
Infecções por Caliciviridae/terapia , Norovirus/genética , Anticorpos de Domínio Único/farmacologia , Sítios de Ligação/genética , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Microscopia Crioeletrônica/métodos , Cristalografia por Raios X/métodos , Epitopos/metabolismo , Gastroenterite/metabolismo , Norovirus/imunologia , Norovirus/patogenicidade , Ligação Proteica/genética , Conformação Proteica , Domínios Proteicos/genética , Anticorpos de Domínio Único/imunologia , Anticorpos de Domínio Único/metabolismo , Vírion/metabolismo
13.
Antioxid Redox Signal ; 33(1): 1-19, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32103677

RESUMO

Aims: Mitochondrial stress and dysfunction within the intestinal epithelium are known to contribute to the pathogenesis of inflammatory bowel disease (IBD). However, the importance of mitophagy during intestinal inflammation remains poorly understood. The primary aim of this study was to investigate how the mitophagy protein BCL2/adenovirus E1B 19 kDa protein-interacting protein 3-like (BNIP3L/NIX) mitigates mitochondrial damage during intestinal inflammation in the hopes that these data will allow us to target mitochondrial health in the intestinal epithelium as an adjunct to immune-based treatment strategies. Results: In the intestinal epithelium of patients with ulcerative colitis, we found that NIX was upregulated and targeted to the mitochondria. We obtained similar findings in wild-type mice undergoing experimental colitis. An increase in NIX expression was found to depend on stabilization of hypoxia-inducible factor-1 alpha (HIF1α), which binds to the Nix promoter region. Using the reactive oxygen species (ROS) scavenger MitoTEMPO, we were able to attenuate disease and inhibit both HIF1α stabilization and subsequent NIX expression, suggesting that mitochondrially derived ROS are crucial to initiating the mitophagic response during intestinal inflammation. We subjected a global Nix-/- mouse to dextran sodium sulfate colitis and found that these mice developed worse disease. In addition, Nix-/- mice were found to exhibit increased mitochondrial mass, likely due to the inability to clear damaged or dysfunctional mitochondria. Innovation: These results demonstrate the importance of mitophagy within the intestinal epithelium during IBD pathogenesis. Conclusion: NIX-mediated mitophagy is required to maintain intestinal homeostasis during inflammation, highlighting the impact of mitochondrial damage on IBD progression.


Assuntos
Gastroenterite/etiologia , Proteínas de Membrana/genética , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Mitofagia/genética , Animais , Antioxidantes/farmacologia , Sítios de Ligação , Biomarcadores , Linhagem Celular Tumoral , Colite/etiologia , Colite/metabolismo , Colite/patologia , Óxidos N-Cíclicos/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Gastroenterite/metabolismo , Gastroenterite/patologia , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Modelos Biológicos , Regiões Promotoras Genéticas , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo , Elementos de Resposta
14.
Antioxid Redox Signal ; 33(5): 354-373, 2020 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-31968991

RESUMO

Significance: Despite their intrinsic cytotoxic properties, mounting evidence indicates that reactive oxygen species (ROS) physiologically produced by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) of epithelial cells (NOX1, dual oxidase [DUOX]2) and phagocytes (NOX2) are critical for innate immune response and homeostasis of the intestinal mucosa. However, dysregulated ROS production could be a driving factor in inflammatory bowel diseases (IBDs). Recent Advances: In addition to NOX2, recent studies have demonstrated that NOX1- and DUOX2-derived ROS can regulate intestinal innate immune defense and homeostasis by impacting many processes, including bacterial virulence, expression of bacteriostatic proteins, epithelial renewal and restitution, and microbiota composition. Moreover, the antibacterial role of DUOX2 is a function conserved in evolution as it has been described in invertebrates, and lower and higher vertebrates. In humans, variants of the NOX2, NOX1, and DUOX2 genes, which are associated with impaired ROS production, have been identified in very early onset IBD, but overexpression of NOX/DUOX, especially DUOX2, has also been described in IBD, suggesting that loss-of-function or excessive activity of the ROS-generating enzymes could contribute to disease progression. Critical Issues: Therapeutic perspectives aiming at targeting NOX/DUOX in IBD should take into account the two sides of NOX/DUOX-derived ROS in intestinal inflammation. Hence, NOX/DUOX inhibitors or ROS inducers should be considered as a function of the disease context. Future Directions: A thorough understanding of the physiological and pathological regulation of NOX/DUOX in the gastrointestinal tract is an absolute pre-requisite for the development of therapeutic strategies that can modulate ROS levels in space and time.


Assuntos
Gastroenterite/etiologia , Gastroenterite/metabolismo , Trato Gastrointestinal/metabolismo , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Biomarcadores , Gerenciamento Clínico , Suscetibilidade a Doenças , Oxidases Duais/genética , Oxidases Duais/metabolismo , Gastroenterite/patologia , Gastroenterite/terapia , Trato Gastrointestinal/patologia , Expressão Gênica , Humanos , Imunidade Inata , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Doenças Inflamatórias Intestinais/terapia
15.
Sci Immunol ; 5(54)2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33443029

RESUMO

Deficiency in interleukin-36R (IL-36R) antagonist caused by loss-of-function mutations in IL-36RN leads to DITRA (deficiency of IL-36 receptor antagonist), a rare inflammatory human disease that belongs to a subgroup of generalized pustular psoriasis (GPP). We report a functional genetic mouse model of DITRA with enhanced IL-36R signaling analogous to that observed in patients with DITRA, which provides new insight into our understanding of the IL-36 family of molecules in regulating barrier integrity across multiple tissues. Humanized DITRA-like mice displayed increased skin inflammation in a preclinical model of psoriasis, and in vivo blockade of IL-36R pathway using anti-human IL-36R antibody ameliorated imiquimod-induced skin pathology as both prophylactic and therapeutic treatments. Deeper characterization of the humanized DITRA-like mice revealed that deregulated IL-36R signaling promoted tissue pathology during intestinal injury and led to impairment in mucosal restoration in the repair phase of chronic dextran sulfate sodium (DSS)-induced colitis. Blockade of IL-36R pathway significantly ameliorated DSS-induced intestinal inflammation and rescued the inability of DITRA-like mice to recover from mucosal damage in vivo. Our results indicate a central role for IL-36 in regulating proinflammatory responses in the skin and epithelial barrier function in the intestine, suggesting a new therapeutic potential for targeting the IL-36R axis in psoriasis and at the later stages of intestinal pathology in inflammatory bowel disease.


Assuntos
Dermatite/etiologia , Dermatite/metabolismo , Gastroenterite/etiologia , Gastroenterite/metabolismo , Receptores de Interleucina-1/metabolismo , Transdução de Sinais , Animais , Biomarcadores , Dermatite/patologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Gastroenterite/patologia , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Pele/metabolismo , Pele/patologia
16.
Cancer Res ; 79(16): 4003-4008, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31362930

RESUMO

Metabolic reprogramming of cancer cells and the tumor microenvironment are emerging as key factors governing tumor growth, metastasis, and response to therapies including immune checkpoint inhibitors. It has been recognized that rapidly proliferating cancer cells, tumor-infiltrating lymphocytes, and vascular endothelial cells compete for oxygen and nutrients. Tumor cells and other cell types in the microenvironment not only compete for nutrients, but they also simultaneously produce immunosuppressive metabolites, leading to immune escape. In addition, commensal microbial metabolites can influence regulatory T cells and inflammation in the intestine, thus playing an essential role in cancer prevention or cancer promotion. In this review, we summarize recent advances on metabolic interactions among various cell types in the tumor microenvironment, with a focus on how these interactions affect tumor immunity. We also discuss the potential role of blood vessel metabolism in regulating immune cell trafficking and activation.


Assuntos
Neoplasias Colorretais/microbiologia , Gastroenterite/metabolismo , Microbioma Gastrointestinal/fisiologia , Linfócitos do Interstício Tumoral/metabolismo , Microambiente Tumoral/imunologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Gastroenterite/patologia , Microbioma Gastrointestinal/imunologia , Glicólise , Humanos , Linfócitos do Interstício Tumoral/patologia , Linfócitos T Reguladores/patologia , Linfócitos T Reguladores/fisiologia , Microambiente Tumoral/fisiologia
17.
Arch Toxicol ; 93(8): 2127-2139, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31309260

RESUMO

Chronic exposure to inorganic arsenic (As) [As(III) + As(V)], which affects millions of people, increases the incidence of some kinds of cancer and other non-carcinogenic pathologies. Although the oral pathway is the main form of exposure, in vivo studies have not been conducted to verify the intestinal toxicity of this metalloid. The aim of this study is to perform an in vivo evaluation of the intestinal toxicity of inorganic As, using female BALB/c mice exposed through drinking water to various concentrations of As(III) (20, 50, and 80 mg/L) for 2 months. An increase was observed in oxygen and/or nitrogen reactive species, and in gene and protein expression of pro-inflammatory cytokines (IL-1ß, IL-2, IL-6) at concentrations equal to or greater than 50 mg/L. These changes were accompanied by a profound remodeling of the intestinal microbial profile in terms of diversity and global composition, which could be at the basis or exacerbate As(III) toxic effects. The histological study showed that there was moderate inflammation of the mucosa and submucosa, accompanied by hyperplasia of crypts at the highest administered dose. In addition, all the treatments with As(III) resulted in a decreased expression of Muc2, which encodes one of the main components of the intestinal layer of mucus. The effects described are compatible with the increased intestinal permeability observed at concentrations equal to or greater than 50 mg/L, indicative of loss of barrier function.


Assuntos
Arsenitos/toxicidade , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Animais , Arsenitos/administração & dosagem , Citocinas/genética , Feminino , Gastroenterite/induzido quimicamente , Gastroenterite/metabolismo , Gastroenterite/patologia , Camundongos Endogâmicos BALB C , Mucina-2/genética , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Testes de Toxicidade Subcrônica
18.
Nutrients ; 11(7)2019 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-31252646

RESUMO

Citrus flavanones, with hesperidin and naringin as the most abundant representatives, have various beneficial effects, including anti-oxidative and anti-inflammatory activities. Evidence also indicates that they may impact the intestinal microbiome and are metabolized by the microbiota as well, thereby affecting their bioavailability. In this review, we provide an overview on the current evidence on the intestinal fate of hesperidin and naringin, their interaction with the gut microbiota, and their effects on intestinal barrier function and intestinal inflammation. These topics will be discussed as they may contribute to gastrointestinal health in various diseases. Evidence shows that hesperidin and naringin are metabolized by intestinal bacteria, mainly in the (proximal) colon, resulting in the formation of their aglycones hesperetin and naringenin and various smaller phenolics. Studies have also shown that citrus flavanones and their metabolites are able to influence the microbiota composition and activity and exert beneficial effects on intestinal barrier function and gastrointestinal inflammation. Although the exact underlying mechanisms of action are not completely clear and more research in human subjects is needed, evidence so far suggests that citrus flavanones as well as their metabolites have the potential to contribute to improved gastrointestinal function and health.


Assuntos
Bactérias/metabolismo , Citrus/metabolismo , Colo/metabolismo , Flavanonas/metabolismo , Frutas/metabolismo , Gastroenterite/prevenção & controle , Microbioma Gastrointestinal , Hesperidina/metabolismo , Absorção Intestinal , Intestino Delgado/metabolismo , Animais , Bactérias/efeitos dos fármacos , Disponibilidade Biológica , Colo/efeitos dos fármacos , Colo/microbiologia , Flavanonas/administração & dosagem , Gastroenterite/metabolismo , Gastroenterite/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Hesperidina/administração & dosagem , Humanos , Absorção Intestinal/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/microbiologia
19.
Immunity ; 50(6): 1365-1379, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31216461

RESUMO

The mammalian intestine is colonized by a wealth of microorganisms-including bacteria, viruses, protozoa, and fungi-that are all integrated into a functional trans-kingdom community. Characterization of the composition of the fungal community-the mycobiota-has advanced further than the much-needed mechanistic studies. Recent findings have revealed roles for the gut mycobiota in the regulation of host immunity and in the development and progression of human diseases of inflammatory origin. We review these findings here while placing them in the context of the current understanding of the pathways and cellular networks that induce local and systemic immune responses to fungi in the gastrointestinal tract. We discuss gaps in knowledge and argue for the importance of considering bacteria-fungal interactions as we aim to define the roles of mycobiota in immune homeostasis and immune-associated pathologies.


Assuntos
Suscetibilidade a Doenças , Gastroenterite/etiologia , Microbioma Gastrointestinal/imunologia , Imunidade , Imunidade Adaptativa , Animais , Suscetibilidade a Doenças/imunologia , Gastroenterite/metabolismo , Homeostase , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata
20.
Trends Mol Med ; 25(6): 464-466, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31109795

RESUMO

Vancomycin-resistant Enterococcus faecium (VRE) is a major cause of nosocomial infections. A new study by McKenney et al. (Cell Host Microbe 2019;25:695-705.e5) reports that VRE undergo a morphotype switch in response to lithocholic acid (LCA) to facilitate gastrointestinal (GI) tract colonization. This metabolic cue is a potential target to decrease VRE colonization and subsequent transmission of antibiotic resistance.


Assuntos
Enterococcus/fisiologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Transdução de Sinais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Ácidos e Sais Biliares/metabolismo , Farmacorresistência Bacteriana , Enterococcus/citologia , Enterococcus/efeitos dos fármacos , Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/fisiologia , Gastroenterite/metabolismo , Gastroenterite/microbiologia , Infecções por Bactérias Gram-Positivas/metabolismo , Infecções por Bactérias Gram-Positivas/microbiologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...