Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Science ; 381(6662): eabq5202, 2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37676943

RESUMO

Kupffer cells (KCs) are localized in liver sinusoids but extend pseudopods to parenchymal cells to maintain their identity and serve as the body's central bacterial filter. Liver cirrhosis drastically alters vascular architecture, but how KCs adapt is unclear. We used a mouse model of liver fibrosis and human tissue to examine immune adaptation. Fibrosis forced KCs to lose contact with parenchymal cells, down-regulating "KC identity," which rendered them incapable of clearing bacteria. Commensals stimulated the recruitment of monocytes through CD44 to a spatially distinct vascular compartment. There, recruited monocytes formed large aggregates of multinucleated cells (syncytia) that expressed phenotypical KC markers and displayed enhanced bacterial capture ability. Syncytia formed via CD36 and were observed in human cirrhosis as a possible antimicrobial defense that evolved with fibrosis.


Assuntos
Infecções Transmitidas por Sangue , Células Gigantes , Células de Kupffer , Cirrose Hepática , Animais , Humanos , Camundongos , Células Gigantes/imunologia , Células Gigantes/microbiologia , Células de Kupffer/imunologia , Células de Kupffer/microbiologia , Cirrose Hepática/imunologia , Cirrose Hepática/microbiologia , Cirrose Hepática/patologia , Infecções Transmitidas por Sangue/imunologia , Modelos Animais de Doenças
2.
Int J Mol Sci ; 24(8)2023 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-37108484

RESUMO

Diet influences the pathogenesis and clinical course of inflammatory bowel disease (IBD). The Mediterranean diet (MD) is linked to reductions in inflammatory biomarkers and alterations in microbial taxa and metabolites associated with health. We aimed to identify features of the gut microbiome that mediate the relationship between the MD and fecal calprotectin (FCP) in ulcerative colitis (UC). Weighted gene co-expression network analysis (WGCNA) was used to identify modules of co-abundant microbial taxa and metabolites correlated with the MD and FCP. The features considered were gut microbial taxa, serum metabolites, dietary components, short-chain fatty acid and bile acid profiles in participants that experienced an increase (n = 13) or decrease in FCP (n = 16) over eight weeks. WGCNA revealed ten modules containing sixteen key features that acted as key mediators between the MD and FCP. Three taxa (Faecalibacterium prausnitzii, Dorea longicatena, Roseburia inulinivorans) and a cluster of four metabolites (benzyl alcohol, 3-hydroxyphenylacetate, 3-4-hydroxyphenylacetate and phenylacetate) demonstrated a strong mediating effect (ACME: -1.23, p = 0.004). This study identified a novel association between diet, inflammation and the gut microbiome, providing new insights into the underlying mechanisms of how a MD may influence IBD. See clinicaltrials.gov (NCT04474561).


Assuntos
Colite Ulcerativa , Dieta Mediterrânea , Doenças Inflamatórias Intestinais , Humanos , Colite Ulcerativa/microbiologia , Doenças Inflamatórias Intestinais/microbiologia , Inflamação/genética , Biomarcadores , Fezes/microbiologia
3.
Mucosal Immunol ; 15(6): 1085-1094, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-36065057

RESUMO

The gut microbiota influences host responses at practically every level, and as research into host-microbe interactions expands, it is not surprising that we are uncovering similar roles for the microbiota at other barrier sites, such as the lung and skin. Using standard laboratory mice to assess host-microbe interactions, or even host intrinsic responses, can be challenging, as slight variations in the microbiota can affect experimental outcomes. When it comes to designing and selecting an appropriate level of microbial diversity and community structure for colonization of our laboratory rodents, we have more choices available to us than ever before. Here we will discuss the different approaches used to modulate microbial complexity that are available to study host-microbe interactions. We will describe how different models have been used to answer distinct biological questions, covering the entire microbial spectrum, from germ-free to wild.


Assuntos
Microbioma Gastrointestinal , Microbiota , Camundongos , Animais , Microbioma Gastrointestinal/fisiologia , Interações entre Hospedeiro e Microrganismos , Mucosa
4.
Cancers (Basel) ; 14(11)2022 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-35681702

RESUMO

The gut microbiota plays a role in shaping overall host health and response to several cancer treatments. Factors, such as diet, exercise, and chemotherapy, can alter the gut microbiota. In the present study, the Alberta Cancer Exercise (ACE) program was investigated as a strategy to favorably modify the gut microbiota of breast cancer survivors who had received chemotherapy. Subsequently, the ability of post-exercise gut microbiota, alone or with prebiotic fiber supplementation, to influence breast cancer outcomes was interrogated using fecal microbiota transplant (FMT) in germ-free mice. While cancer survivors experienced little gut microbial change following ACE, in the mice, tumor volume trended consistently lower over time in mice colonized with post-exercise compared to pre-exercise microbiota with significant differences on days 16 and 22. Beta diversity analysis revealed that EO771 breast tumor cell injection and Paclitaxel chemotherapy altered the gut microbial communities in mice. Enrichment of potentially protective microbes was found in post-exercise microbiota groups. Tumors of mice colonized with post-exercise microbiota exhibited more favorable cytokine profiles, including decreased vascular endothelial growth factor (VEGF) levels. Beneficial microbial and molecular outcomes were augmented with prebiotic supplementation. Exercise and prebiotic fiber demonstrated adjuvant action, potentially via an enhanced anti-tumor immune response modulated by advantageous gut microbial shifts.

5.
J Agric Food Chem ; 70(16): 5005-5014, 2022 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-35420414

RESUMO

Thearubigins, polymers of tea catechins, account for more than 20% of the black tea polyphenols and have been reported to be the active components in black tea. However, the chemical structures and underlying mechanisms regarding how the thearubigins, being poorly bioavailable, generate in vivo health benefits are still largely unknown. Using germ-free and specific pathogen-free husbandry conditions combined with LC/MS-based nontargeted and targeted metabolomic analyses, we investigated the role of intestinal bacteria in thearubigin metabolism. Theaflavins and theasinensins were identified as the major microbial metabolites of thearubigins, suggesting that these molecules are the building units for the complex thearubigins. To further confirm this, thearubigin depolymerization was done using menthofuran in an acidic condition. Menthofuran-conjugated theaflavins, theasinensins, and catechins as well as their free forms were detected as the major degradation products of thearubigins. This indicated that theaflavins and theasinensins could be further polymerized through B-type proanthocyanidin linkages. Furthermore, four microbial degradation products were able to be detected in urine samples, suggesting that they can be absorbed into the circulatory system. Using the combination of microbial degradation, metabolomics, and chemical degradation, our results demonstrate that thearubigins are the complex polymers of theaflavins, theasinensins, and catechins and can be metabolized by gut microbiota to their corresponding bioactive and bioavailable smaller molecular metabolites.


Assuntos
Camellia sinensis , Catequina , Microbioma Gastrointestinal , Antioxidantes/análise , Camellia sinensis/química , Catequina/química , Espectrometria de Massas/métodos , Polímeros , Polifenóis/química , Chá/química
6.
Microbiome ; 9(1): 186, 2021 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-34517928

RESUMO

BACKGROUND: Studies on the inhibition of inflammation by infection with helminth parasites have, until recently, overlooked a key determinant of health: the gut microbiota. Infection with helminths evokes changes in the composition of their host's microbiota: one outcome of which is an altered metabolome (e.g., levels of short-chain fatty acids (SCFAs)) in the gut lumen. The functional implications of helminth-evoked changes in the enteric microbiome (composition and metabolites) are poorly understood and are explored with respect to controlling enteric inflammation. METHODS: Antibiotic-treated wild-type, germ-free (GF) and free fatty-acid receptor-2 (ffar2) deficient mice were infected with the tapeworm Hymenolepis diminuta, then challenged with DNBS-colitis and disease severity and gut expression of the il-10 receptor-α and SCFA receptors/transporters assessed 3 days later. Gut bacteria composition was assessed by 16 s rRNA sequencing and SCFAs were measured. Other studies assessed the ability of feces or a bacteria-free fecal filtrate from H. diminuta-infected mice to inhibit colitis. RESULTS: Protection against disease by infection with H. diminuta was abrogated by antibiotic treatment and was not observed in GF-mice. Bacterial community profiling revealed an increase in variants belonging to the families Lachnospiraceae and Clostridium cluster XIVa in mice 8 days post-infection with H. diminuta, and the transfer of feces from these mice suppressed DNBS-colitis in GF-mice. Mice treated with a bacteria-free filtrate of feces from H. diminuta-infected mice were protected from DNBS-colitis. Metabolomic analysis revealed increased acetate and butyrate (both or which can reduce colitis) in feces from H. diminuta-infected mice, but not from antibiotic-treated H. diminuta-infected mice. H. diminuta-induced protection against DNBS-colitis was not observed in ffar2-/- mice. Immunologically, anti-il-10 antibodies inhibited the anti-colitic effect of H. diminuta-infection. Analyses of epithelial cell lines, colonoids, and colon segments uncovered reciprocity between butyrate and il-10 in the induction of the il-10-receptor and butyrate transporters. CONCLUSION: Having defined a feed-forward signaling loop between il-10 and butyrate following infection with H. diminuta, this study identifies the gut microbiome as a critical component of the anti-colitic effect of this helminth therapy. We suggest that any intention-to-treat with helminth therapy should be based on the characterization of the patient's immunological and microbiological response to the helminth.


Assuntos
Colite , Helmintos , Himenolepíase , Animais , Bactérias/genética , Colite/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C
7.
J Nutr ; 151(6): 1426-1435, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33694368

RESUMO

BACKGROUND: Oat has been widely accepted as a key food for human health. It is becoming increasingly evident that individual differences in metabolism determine how different individuals benefit from diet. Both host genetics and the gut microbiota play important roles on the metabolism and function of dietary compounds. OBJECTIVES: To investigate the mechanism of individual variations in response to whole-grain (WG) oat intake. METHODS: We used the combination of in vitro incubation assays with human gut microbiota, mouse and human S9 fractions, chemical analyses, germ-free (GF) mice, 16S rRNA sequencing, gnotobiotic techniques, and a human feeding study. RESULTS: Avenanthramides (AVAs), the signature bioactive polyphenols of WG oat, were not metabolized into their dihydro forms, dihydro-AVAs (DH-AVAs), by both human and mouse S9 fractions. DH-AVAs were detected in the colon and the distal regions but not in the proximal and middle regions of the perfused mouse intestine, and were in specific pathogen-free (SPF) mice but not in GF mice. A kinetic study of humans fed oat bran showed that DH-AVAs reached their maximal concentrations at much later time points than their corresponding AVAs (10.0-15.0 hours vs. 4.0-4.5 hours, respectively). We observed interindividual variations in the metabolism of AVAs to DH-AVAs in humans. Faecalibacterium prausnitzii was identified as the individual bacterium to metabolize AVAs to DH-AVAs by 16S rRNA sequencing analysis. Moreover, as opposed to GF mice, F. prausnitzii-monocolonized mice were able to metabolize AVAs to DH-AVAs. CONCLUSIONS: These findings demonstrate that the presence of intestinal F. prausnitzii is indispensable for proper metabolism of AVAs in both humans and mice. We propose that the abundance of F. prausnitzii can be used to subcategorize individuals into AVA metabolizers and nonmetabolizers after WG oat intake. This study was registered at clinicaltrials.gov as NCT04335435.


Assuntos
Avena , Faecalibacterium prausnitzii , Microbioma Gastrointestinal , ortoaminobenzoatos/metabolismo , Animais , Avena/química , Dieta , Humanos , Camundongos , RNA Ribossômico 16S/genética
8.
Mol Nutr Food Res ; 65(4): e2000887, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33381889

RESUMO

SCOPE: This study is to determine the in vivo efficacy of black tea theaflavin (TF) to detoxify two metabolic toxins, ammonia and methylglyoxal (MGO), in mice METHODS AND RESULTS: Under in vitro conditions, TF is able to react with ammonia, MGO, and hydrogen peroxide to produce its aminated, MGO conjugated, and oxidized products, respectively. In TF-treated mice, the aminated TF, the MGO conjugates of TF and aminated TF, and the oxidized TF are searched using LC-MS/MS. The results provide the first in vivo evidence that the unabsorbed TF is able to trap ammonia to form the aminated TF; furthermore, both TF and the aminated TF have the capacity to trap MGO to generate the corresponding mono-MGO conjugates. Moreover, TF is oxidized to dehydrotheaflavin, which underwent further amination in the gut. By exposing TF to germ-free (GF) mice and conventionalized mice (GF mice colonized with specific-pathogen-free microbiota), the gut microbiota is demonstrated to facilitate the amination and MGO conjugation of TF. CONCLUSION: TF has the capacity to remove the endogenous metabolic toxins through oxidation, amination, and MGO conjugation in the intestinal tract, which can potentially explain why TF still generates in vivo efficacy while showing a poor systematic bioavailability.


Assuntos
Amônia/farmacocinética , Biflavonoides/farmacologia , Catequina/farmacologia , Aldeído Pirúvico/farmacologia , Chá/química , Amônia/química , Animais , Biflavonoides/química , Biflavonoides/farmacocinética , Catequina/química , Catequina/farmacocinética , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Camundongos Endogâmicos , Oxirredução , Aldeído Pirúvico/química , Organismos Livres de Patógenos Específicos , Toxinas Biológicas/farmacocinética
9.
Curr Opin Microbiol ; 59: 34-41, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32846371

RESUMO

The intestinal microbiota is comprises a diverse community of micro-organisms that interact with many host processes. Innate immune responses to the gut microbiota are of particular importance as they influence many other downstream responses. This fascinating host-microbe crosstalk is a rapidly expanding field of study; thus, it is critical to ensure reproducibility between studies and applicability to human clinical trials through standardization of experiments. We discuss here recent advances in the field including the spectrum of colonization statuses available, the critical importance of colonization timing, the dynamics of the microbial community, and the required housing of animals, as they pertain to appropriate experimental control and design.


Assuntos
Microbioma Gastrointestinal , Imunidade Inata , Projetos de Pesquisa , Animais , Microbioma Gastrointestinal/imunologia , Abrigo para Animais/normas , Microbiota/imunologia , Reprodutibilidade dos Testes , Projetos de Pesquisa/normas
10.
Sci Rep ; 10(1): 6156, 2020 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-32273533

RESUMO

Antigen (Ag)-specific tolerization prevents type 1 diabetes (T1D) in non-obese diabetic (NOD) mice but proved less effective in humans. Several auto-Ags are fundamental to disease development, suggesting T1D etiology is heterogeneous and may limit the effectiveness of Ag-specific therapies to distinct disease endotypes. Colonization factor antigen I (CFA/I) fimbriae from Escherichia coli can inhibit autoimmune diseases in murine models by inducing bystander tolerance. To test if Ag-independent stimulation of regulatory T cells (Tregs) can prevent T1D onset, groups of NOD mice were orally treated with Lactococcus lactis (LL) expressing CFA/I. LL-CFA/I treatment beginning at 6 weeks of age reduced disease incidence by 50% (p < 0.05) and increased splenic Tregs producing both IL-10 and IFN-γ 8-fold (p < 0.005) compared to LL-vehicle treated controls. To further describe the role of these Tregs in preventing T1D, protective phenotypes were examined at different time-points. LL-CFA/I treatment suppressed splenic TNF-α+CD8+ T cells 6-fold at 11 weeks (p < 0.005) and promoted a distinct microbiome. At 17 weeks, IFN-γ+CD4+ T cells were suppressed 10-fold (p < 0.005), and at 30 weeks, pancreatic Tbet+CD4+ T cells were suppressed (p < 0.05). These results show oral delivery of modified commensal organisms, such as LL-CFA/I, may be harnessed to restrict Th1 cell-mediated immunity and protect against T1D.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Proteínas de Fímbrias/uso terapêutico , Administração Oral , Animais , Modelos Animais de Doenças , Feminino , Proteínas de Fímbrias/administração & dosagem , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , Linfócitos T Reguladores/fisiologia
11.
Free Radic Biol Med ; 131: 332-344, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30578921

RESUMO

The in vivo mechanism of tea polyphenol-mediated prevention of many chronic diseases is still largely unknown. Studies have shown that accumulation of toxic reactive cellular metabolites, such as ammonia and reactive carbonyl species (RCS), is one of the causing factors to the development of many chronic diseases. In this study, we investigated the in vivo interaction between (-)-epigallocatechin-3-gallate (EGCG), the most abundant polyphenol in tea leaves, and ammonia and RCS. We found that EGCG could be oxidized to EGCG quinone in mice, and then rapidly react with ammonia to generate the aminated EGCG metabolite, 4'-NH2-EGCG. Both EGCG and its aminated metabolite could further scavenge RCS, such as methylglyoxal (MGO), malondialdehyde (MDA), and trans-4-hydroxy-2-nonenal (4-HNE), to produce the RCS conjugates of EGCG and the aminated EGCG. Both the aminated and the RCS conjugated metabolites of EGCG were detected in human after drinking four cups of green tea per day. By comparing the levels of the aminated and the RCS conjugated metabolites in EGCG exposed germ-free (GF) mice and specific-pathogen-free (SPF) mice, we demonstrated that gut microbiota facilitate the formation of the aminated metabolite of EGCG, the RCS conjugates of EGCG, and the RCS conjugates of the aminated EGCG. By comparing the trapping capacities of EGCG and its aminated metabolite under aerobic and anaerobic conditions, we found that oxygen is not essential for the trapping of reactive species by EGCG and 4'-NH2-EGCG suggesting that EGCG and its aminated metabolite could scavenge RCS in the GI track and in the circulation system. Altogether, this study provides in vivo evidences that EGCG has the capacity to scavenge toxic reactive metabolic wastes. This finding opens a new window to understand the underlying mechanisms by which drinking tea could prevent the development of chronic diseases.


Assuntos
Aldeídos/metabolismo , Catequina/análogos & derivados , Sequestradores de Radicais Livres/metabolismo , Malondialdeído/metabolismo , Aldeído Pirúvico/metabolismo , Chá/metabolismo , Aminação , Amônia/metabolismo , Animais , Catequina/metabolismo , Microbioma Gastrointestinal/fisiologia , Vida Livre de Germes , Células HCT116 , Células HT29 , Humanos , Camundongos , Oxirredução , Quinonas/metabolismo , Desintoxicação por Sorção/métodos
12.
Mucosal Immunol ; 11(5): 1329-1341, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29875400

RESUMO

Intestinal epithelial Na+/H+ exchange facilitated by the apical NHE3 (Slc9a3) is a highly regulated process inhibited by intestinal pathogens and in inflammatory bowel diseases. NHE3-/- mice develop spontaneous, bacterially mediated colitis, and IBD-like dysbiosis. Disruption of epithelial Na+/H+ exchange in IBD may thus represent a host response contributing to the altered gut microbial ecology, and may play a pivotal role in modulating the severity of inflammation in a microbiome-dependent manner. To test whether microbiome fostered in an NHE3-deficient environment is able to drive mucosal immune responses affecting the onset or severity of colitis, we performed a series of cohousing experiments and fecal microbiome transplants into germ-free Rag-deficient or IL-10-/- mice. We determined that in the settings where the microbiome of NHE3-deficient mice was stably engrafted in the recipient host, it was able accelerate the onset and amplify severity of experimental colitis. NHE3-deficiency was characterized by the reduction in pH-sensitive butyrate-producing Firmicutes families Lachnospiraceae and Ruminococcaceae (Clostridia clusters IV and XIVa), with an expansion of inflammation-associated Bacteroidaceae. We conclude that the microbiome fostered by impaired epithelial Na+/H+ exchange enhances the onset and severity of colitis through disruption of the gut microbial ecology.


Assuntos
Colite/metabolismo , Disbiose/metabolismo , Microbioma Gastrointestinal/imunologia , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Bacteroidaceae/imunologia , Disbiose/imunologia , Disbiose/microbiologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Firmicutes/imunologia , Vida Livre de Germes , Concentração de Íons de Hidrogênio , Imunidade/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/microbiologia , Interleucina-10/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Trocador 3 de Sódio-Hidrogênio/metabolismo
13.
Carcinogenesis ; 39(8): 1068-1078, 2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-29846515

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer death in the United States yet data are scant regarding host factors influencing pancreatic carcinogenesis. Increasing evidence support the role of the host microbiota in carcinogenesis but its role in PDAC is not well established. Herein, we report that antibiotic-mediated microbial depletion of KrasG12D/PTENlox/+ mice showed a decreased proportion of poorly differentiated tumors compared to microbiota-intact KrasG12D/PTENlox/+ mice. Subsequent 16S rRNA PCR showed that ~50% of KrasG12D/PTENlox/+ mice with PDAC harbored intrapancreatic bacteria. To determine if a similar observation in humans correlates with presence of PDAC, benign and malignant human pancreatic surgical specimens demonstrated a microbiota by 16S bacterial sequencing and culture confirmation. However, the microbial composition did not differentiate PDAC from non-PDAC tissue. Furthermore, murine pancreas did not naturally acquire a pancreatic microbiota, as germ-free mice transferred to specific pathogen-free housing failed to acquire intrapancreatic bacteria over time, which was not augmented by a murine model of colitis. Finally, antibiotic-mediated microbial depletion of Nod-SCID mice, compared to microbiota-intact, showed increased time to PDAC xenograft formation, smaller tumors, and attenuated growth. Interestingly, both xenograft cohorts were devoid of intratumoral bacteria by 16S rRNA PCR, suggesting that intrapancreatic/intratumoral microbiota is not the sole driver of PDAC acceleration. Xenografts from microbiota-intact mice demonstrated innate immune suppression by immunohistochemistry and differential regulation of oncogenic pathways as determined by RNA sequencing. Our work supports a long-distance role of the intestinal microbiota on PDAC progression and opens new research avenues regarding pancreatic carcinogenesis.


Assuntos
Carcinogênese/imunologia , Carcinoma Ductal Pancreático/imunologia , Microbioma Gastrointestinal/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Neoplasias Pancreáticas/imunologia , Adulto , Idoso , Animais , Antibacterianos/administração & dosagem , Bactérias/efeitos dos fármacos , Bactérias/genética , Bactérias/isolamento & purificação , Carcinogênese/efeitos dos fármacos , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/cirurgia , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Vida Livre de Germes , Interações entre Hospedeiro e Microrganismos/efeitos dos fármacos , Humanos , Intestinos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Pessoa de Meia-Idade , Pâncreas/microbiologia , Pâncreas/patologia , Pâncreas/cirurgia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/cirurgia , RNA Ribossômico 16S/isolamento & purificação , Ensaios Antitumorais Modelo de Xenoenxerto
14.
JCI Insight ; 2(11)2017 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-28570279

RESUMO

Intestinal tuft cells are a rare, poorly understood cell type recently shown to be a critical mediator of type 2 immune response to helminth infection. Here, we present advances in segmentation algorithms and analytical tools for multiplex immunofluorescence (MxIF), a platform that enables iterative staining of over 60 antibodies on a single tissue section. These refinements have enabled a comprehensive analysis of tuft cell number, distribution, and protein expression profiles as a function of anatomical location and physiological perturbations. Based solely on DCLK1 immunoreactivity, tuft cell numbers were similar throughout the mouse small intestine and colon. However, multiple subsets of tuft cells were uncovered when protein coexpression signatures were examined, including two new intestinal tuft cell markers, Hopx and EGFR phosphotyrosine 1068. Furthermore, we identified dynamic changes in tuft cell number, composition, and protein expression associated with fasting and refeeding and after introduction of microbiota to germ-free mice. These studies provide a foundational framework for future studies of intestinal tuft cell regulation and demonstrate the utility of our improved MxIF computational methods and workflow for understanding cellular heterogeneity in complex tissues in normal and disease states.

15.
Nutr Neurosci ; 19(8): 337-345, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26086200

RESUMO

OBJECTIVES: Western-style diets high in saturated fat and refined carbohydrate have been shown to alter gut microbiota as well as being associated with altered behaviour and learning ability. The objective of this study was to determine the effects of short-term intake of a Western-style diet on intestinal cytokine expression, tryptophan metabolism, and levels of neurotransmitters in the brain. METHODS: At 7 weeks of age, 129S1/SvImJ mice were placed on a standard chow or Western-style diet (fat 33%, refined carbohydrates 49%) for 3 weeks. Anxiety-like behaviour was assessed by the latency to step-down test and exploration assessed in a Barnes maze. Neurotransmitter levels in forebrains were analysed by high-pressure liquid chromatography. Liver metabolism was examined by 1H nuclear magnetic resonance (NMR). Cytokine expression in the intestine was measured using MesoScale discovery platform. mRNA levels of tryptophan hydroxylase (Tph) and indoleamine 2,3-dioxygenase (IDO) in the brain and intestine were measured using qPCR. RESULTS: Results showed that mice fed the Western diet displayed reduced exploratory and anxiety-like behaviour. Anxiolytic effects correlated with increased hippocampal brain-derived neurotrophic factor (BDNF) and tryptophan levels. Brain serotonin was not altered. These changes were associated with reduced expression of small intestinal indoleamine 2,3-dioxygenase, a tryptophan-processing enzyme. Western diet-fed mice exhibited low-grade systemic and intestinal inflammation along with altered liver metabolic profiles. DISCUSSION: In conclusion, diets high in fat and refined sugar are associated with increased levels of brain BDNF and tryptophan and decreased exploratory and anxiety-like behaviour. These behavioural changes correlated with altered intestinal tryptophan metabolism and liver metabolic profiles.


Assuntos
Ansiedade/etiologia , Dieta Ocidental/efeitos adversos , Modelos Animais de Doenças , Mucosa Intestinal/metabolismo , Doenças Metabólicas/etiologia , Prosencéfalo/metabolismo , Triptofano/metabolismo , Animais , Ansiedade/imunologia , Ansiedade/metabolismo , Comportamento Animal , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Citocinas/metabolismo , Comportamento Exploratório , Regulação Enzimológica da Expressão Gênica , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Mucosa Intestinal/enzimologia , Mucosa Intestinal/imunologia , Intestino Delgado/enzimologia , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Fígado/imunologia , Fígado/metabolismo , Masculino , Doenças Metabólicas/imunologia , Doenças Metabólicas/metabolismo , Camundongos da Linhagem 129 , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/enzimologia , Neurônios/imunologia , Neurônios/metabolismo , Prosencéfalo/enzimologia , Prosencéfalo/imunologia , Organismos Livres de Patógenos Específicos , Triptofano Hidroxilase/genética , Triptofano Hidroxilase/metabolismo
16.
Cell Mol Gastroenterol Hepatol ; 1(1): 28-40, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25729763

RESUMO

The concept that the intestinal microbiota modulates numerous physiological processes including immune development and function, nutrition and metabolism as well as pathogen exclusion is relatively well established in the scientific community. The molecular mechanisms driving these various effects and the events leading to the establishment of a "healthy" microbiome are slowly emerging. The objective of this review is to bring into focus important aspects of microbial/host interactions in the intestine and to discuss key molecular mechanisms controlling health and disease states. We will discuss recent evidence on how microbes interact with the host and one another and their impact on intestinal homeostasis.

17.
Cell Metab ; 20(6): 937-8, 2014 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-25470545

RESUMO

Obesity and the associated state of subchronic inflammation are risk factors for numerous pathologies, including carcinogenesis. Recently, Schulz et al. (2014) demonstrated that high-fat diet-induced intestinal dysbiosis promotes cancer development in K-ras(G12Dint) mice without inducing obesity or mucosal inflammation, positioning microbial activities as a central component of diet-induced carcinogenesis.


Assuntos
Carcinogênese/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Gorduras na Dieta/efeitos adversos , Disbiose/induzido quimicamente , Disbiose/microbiologia , Neoplasias Intestinais/microbiologia , Obesidade , Animais
18.
Psychoneuroendocrinology ; 38(9): 1738-47, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23566632

RESUMO

Modulation of the gut microbiota with diet and probiotic bacteria can restore intestinal homeostasis in inflammatory conditions and alter behavior via the gut-brain axis. The purpose of this study was to determine whether the modulatory effects of probiotics differ depending on diet and mouse genotype. At weaning, wild type (WT) and IL-10 deficient (IL-10(-/-)) 129/SvEv mice were placed on a standard mouse chow or a Western-style diet (fat 33%, refined carbohydrate 49%)±Lactobacillus helveticus ROO52 (10(9)cfu/d) for 21 days. Animal weight and food eaten were monitored weekly. Intestinal immune function was analysed for cytokine expression using the Meso Scale Discovery platform. Spatial memory and anxiety-like behavior was assessed in a Barnes maze. Terminal restriction fragment length polymorphism (TRFLP) was used to analyze the fecal microbiota. Both WT and IL-10(-/-) mice on a Western diet had increased weight gain along with changes in gut microbiota and cytokine expression and altered anxiety-like behavior. The ability of L. helveticus to modulate these factors was genotype- and diet-dependent. Anxiety-like behavior and memory were negatively affected by Western-style diet depending on inflammatory state, but this change was prevented with L. helveticus administration. However, probiotics alone decreased anxiety-like behavior in WT mice on a chow diet. Mice on the Western diet had decreased inflammation and fecal corticosterone, but these markers did not correlate with changes in behavior. Analysis of bacterial phyla from WT and IL-10(-/-)mice showed discrete clustering of the groups to be associated with both diet and probiotic supplementation, with the diet-induced shift normalized to some degree by L. helveticus. These findings suggest that the type of diet consumed by the host and the presence or absence of active inflammation may significantly alter the ability of probiotics to modulate host physiological function.


Assuntos
Ração Animal , Ansiedade/prevenção & controle , Colite/prevenção & controle , Inflamação/prevenção & controle , Intestinos/microbiologia , Lactobacillus helveticus , Transtornos da Memória/prevenção & controle , Microbiota/fisiologia , Probióticos/uso terapêutico , Animais , Ansiedade/etiologia , Colite/etiologia , Colite/microbiologia , Colite/patologia , Cortisona/análise , Citocinas/metabolismo , Ácidos Graxos/análise , Fezes/química , Conteúdo Gastrointestinal/química , Genótipo , Hipocampo/patologia , Inflamação/etiologia , Interleucina-10/deficiência , Interleucina-10/genética , Intestinos/química , Intestinos/patologia , Lactobacillus helveticus/fisiologia , Aprendizagem em Labirinto , Transtornos da Memória/etiologia , Camundongos , Microbiota/genética , Polimorfismo de Fragmento de Restrição , Probióticos/toxicidade , Prosencéfalo/patologia , Organismos Livres de Patógenos Específicos , Aumento de Peso
19.
Cell Microbiol ; 14(4): 447-59, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22212348

RESUMO

Both pathogenic and commensal strains of Escherichia coli colonize the human intestinal tract. Pathogenic strains differ only in the expression of virulence factors, many of which comprise a type III secretion system (TTSS). Little is known regarding the effect of E. coli on the intestinal epithelial response to the secretagogues that drive ion secretion, despite its importance in causing clinically significant diarrhoea. Using Ussing chambers to measure electrogenic ion transport of T84 intestinal epithelial cell monolayers, we found that all strains of E. coli tested (pathogenic, commensal, probiotic and lab strain) significantly reduced cAMP-dependent ion secretion after 4-8 h exposure. Enteropathogenic E. coli mutants lacking a functional TTSS caused similar hyposecretion while not causing significant apoptosis (as shown by caspase-3 cleavage) or necrosis (lactate dehydrogenase release), as did the commensal strain F18, indicating that epithelial cell death was not the cause of hyposecretion. Enteropathogenic E. coli and the TTSS mutant significantly reduced cell surface expression of the apical anion channel, cystic fibrosis transmembrane conductance regulator, which is likely the mechanism behind the pathogen-induced hyposecretion. However, F18 did not cause cystic fibrosis transmembrane conductance regulator mislocalization and the commensal-induced mechanism remains unclear.


Assuntos
Sistemas de Secreção Bacterianos , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/metabolismo , Infecções por Escherichia coli/metabolismo , Morte Celular , Linhagem Celular , Membrana Celular/metabolismo , Colforsina/análogos & derivados , Colforsina/farmacologia , AMP Cíclico/metabolismo , Diarreia/microbiologia , Escherichia coli Êntero-Hemorrágica/efeitos dos fármacos , Escherichia coli Êntero-Hemorrágica/patogenicidade , Escherichia coli Enteropatogênica/efeitos dos fármacos , Células Epiteliais/microbiologia , Infecções por Escherichia coli/microbiologia , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Transporte Proteico , Fatores de Tempo , Fatores de Virulência/metabolismo , Canais de Ânion Dependentes de Voltagem/metabolismo
20.
Am J Physiol Gastrointest Liver Physiol ; 298(6): G807-19, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20299599

RESUMO

The intestinal tract is a diverse microenvironment where more than 500 species of bacteria thrive. A single layer of epithelium is all that separates these commensal microorganisms and pathogens from the underlying immune cells, and thus epithelial barrier function is a key component in the arsenal of defense mechanisms required to prevent infection and inflammation. The epithelial barrier consists of a dense mucous layer containing secretory IgA and antimicrobial peptides as well as dynamic junctional complexes that regulate permeability between cells. Probiotics are live microorganisms that confer benefit to the host and that have been suggested to ameliorate or prevent diseases including antibiotic-associated diarrhea, irritable bowel syndrome, and inflammatory bowel disease. Probiotics likely function through enhancement of barrier function, immunomodulation, and competitive adherence to the mucus and epithelium. This review summarizes the evidence about effects of the many available probiotics with an emphasis on intestinal barrier function and the mechanisms affected by probiotics.


Assuntos
Bactérias/classificação , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiologia , Probióticos , Animais , Fenômenos Fisiológicos Bacterianos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...