Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Gastroenterol Hepatol ; 17(1): 131-148, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37739064

RESUMO

BACKGROUND & AIMS: Altered plasma acylcarnitine levels are well-known biomarkers for a variety of mitochondrial fatty acid oxidation disorders and can be used as an alternative energy source for the intestinal epithelium when short-chain fatty acids are low. These membrane-permeable fatty acid intermediates are excreted into the gut lumen via bile and are increased in the feces of patients with inflammatory bowel disease (IBD). METHODS: Herein, based on studies in human subjects, animal models, and bacterial cultures, we show a strong positive correlation between fecal carnitine and acylcarnitines and the abundance of Enterobacteriaceae in IBD where they can be consumed by bacteria both in vitro and in vivo. RESULTS: Carnitine metabolism promotes the growth of Escherichia coli via anaerobic respiration dependent on the cai operon, and acetylcarnitine dietary supplementation increases fecal carnitine levels with enhanced intestinal colonization of the enteric pathogen Citrobacter rodentium. CONCLUSIONS: In total, these results indicate that the increased luminal concentrations of carnitine and acylcarnitines in patients with IBD may promote the expansion of pathobionts belonging to the Enterobacteriaceae family, thereby contributing to disease pathogenesis.


Assuntos
Enterobacteriaceae , Doenças Inflamatórias Intestinais , Animais , Humanos , Enterobacteriaceae/metabolismo , Disbiose , Doenças Inflamatórias Intestinais/microbiologia , Carnitina/metabolismo , Ácidos Graxos/metabolismo , Escherichia coli , Biomarcadores
2.
Hepatology ; 78(6): 1843-1857, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37222264

RESUMO

BACKGROUND AND AIMS: There is great interest in identifying microbiome features as reliable noninvasive diagnostic and/or prognostic biomarkers for non-cirrhotic NASH fibrosis. Several cross-sectional studies have reported gut microbiome features associated with advanced NASH fibrosis and cirrhosis, where the most prominent features are associated with cirrhosis. However, no large, prospectively collected data exist establishing microbiome features that discern non-cirrhotic NASH fibrosis, integrate the fecal metabolome as disease biomarkers, and are unconfounded by BMI and age. APPROACH AND RESULTS: Results from shotgun metagenomic sequencing performed on fecal samples prospectively collected from 279 US patients with biopsy-proven NASH (F1-F3 fibrosis) enrolled in the REGENERATE I303 study were compared to those from 3 healthy control cohorts and integrated with the absolute quantification of fecal bile acids. Microbiota beta-diversity was different, and BMI- and age-adjusted logistic regression identified 12 NASH-associated species. Random forest prediction models resulted in an AUC of 0.75-0.81 in a receiver operator characteristic analysis. In addition, specific fecal bile acids were significantly lower in NASH and correlated with plasma C4 levels. Microbial gene abundance analysis revealed 127 genes increased in controls, many involving protein synthesis, whereas 362 genes were increased in NASH many involving bacterial environmental responses (false discovery rate < 0.01). Finally, we provide evidence that fecal bile acid levels may be a better discriminator of non-cirrhotic NASH versus health than either plasma bile acids or gut microbiome features. CONCLUSIONS: These results may have value as a set of baseline characteristics of non-cirrhotic NASH against which therapeutic interventions to prevent cirrhosis can be compared and microbiome-based diagnostic biomarkers identified.


Assuntos
Microbioma Gastrointestinal , Hepatopatia Gordurosa não Alcoólica , Humanos , Hepatopatia Gordurosa não Alcoólica/complicações , Estudos Transversais , Cirrose Hepática/complicações , Fibrose , Ácidos e Sais Biliares , Fezes/microbiologia , Biomarcadores
3.
Cell Rep ; 41(11): 111809, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36516747

RESUMO

The gut microbiota influences acetylation on host histones by fermenting dietary fiber into butyrate. Although butyrate could promote histone acetylation by inhibiting histone deacetylases, it may also undergo oxidation to acetyl-coenzyme A (CoA), a necessary cofactor for histone acetyltransferases. Here, we find that epithelial cells from germ-free mice harbor a loss of histone H4 acetylation across the genome except at promoter regions. Using stable isotope tracing in vivo with 13C-labeled fiber, we demonstrate that the microbiota supplies carbon for histone acetylation. Subsequent metabolomic profiling revealed hundreds of labeled molecules and supported a microbial contribution to host fatty acid metabolism, which declined in response to colitis and correlated with reduced expression of genes involved in fatty acid oxidation. These results illuminate the flow of carbon from the diet to the host via the microbiota, disruptions to which may affect energy homeostasis in the distal gut and contribute to the development of colitis.


Assuntos
Colite , Microbiota , Camundongos , Animais , Acetilação , Histonas/metabolismo , Histona Acetiltransferases/metabolismo , Isótopos/metabolismo , Carbono/metabolismo , Butiratos , Ácidos Graxos
4.
Gastroenterology ; 162(3): 743-756, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34774538

RESUMO

BACKGROUND & AIMS: Epidemiologic and murine studies suggest that dietary emulsifiers promote development of diseases associated with microbiota dysbiosis. Although the detrimental impact of these compounds on the intestinal microbiota and intestinal health have been demonstrated in animal and in vitro models, impact of these food additives in healthy humans remains poorly characterized. METHODS: To examine this notion in humans, we performed a double-blind controlled-feeding study of the ubiquitous synthetic emulsifier carboxymethylcellulose (CMC) in which healthy adults consumed only emulsifier-free diets (n = 9) or an identical diet enriched with 15 g per day of CMC (n = 7) for 11 days. RESULTS: Relative to control subjects, CMC consumption modestly increased postprandial abdominal discomfort and perturbed gut microbiota composition in a way that reduced its diversity. Moreover, CMC-fed subjects exhibited changes in the fecal metabolome, particularly reductions in short-chain fatty acids and free amino acids. Furthermore, we identified 2 subjects consuming CMC who exhibited increased microbiota encroachment into the normally sterile inner mucus layer, a central feature of gut inflammation, as well as stark alterations in microbiota composition. CONCLUSIONS: These results support the notion that the broad use of CMC in processed foods may be contributing to increased prevalence of an array of chronic inflammatory diseases by altering the gut microbiome and metabolome (ClinicalTrials.gov, number NCT03440229).


Assuntos
Carboximetilcelulose Sódica/efeitos adversos , Dieta/efeitos adversos , Emulsificantes/efeitos adversos , Microbioma Gastrointestinal/efeitos dos fármacos , Metaboloma/efeitos dos fármacos , Animais , Método Duplo-Cego , Disbiose/etiologia , Fezes , Feminino , Voluntários Saudáveis , Humanos , Masculino , Camundongos
5.
Gut Microbes ; 13(1): 1987781, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34747331

RESUMO

Most studies examining correlations between the gut microbiota and disease states focus on fecal samples due to ease of collection, yet there are distinct differences when compared to samples collected from the colonic mucosa. Although fecal microbiota has been reported to be altered in cirrhosis, correlation with mucosal microbiota characterized via rectal swab has not been previously described in this patient population. We conducted a cross-sectional analysis using 39 stool and 39 rectal swabs from adult patients with cirrhosis of different etiologies and performed shotgun metagenomic sequencing. Bacterial growth studies were performed with Escherichia coli. Two asaccharolytic bacterial taxa, Finegoldia magna and Porphyromonas asaccharolytica, were increased in rectal swabs relative to stool (FDR < 0.01). Genomic analysis of the microbiome revealed 58 genes and 16 pathways that differed between stool and rectal swabs (FDR < 0.05), where rectal swabs were enriched for pathways associated with protein synthesis and cellular proliferation but decreased in carbohydrate metabolism. Although no features in the fecal microbiome differentiated cirrhosis etiologies, the mucosal microbiome revealed decreased abundances of E. coli and Enterobacteriaceae in alcohol-related cirrhosis relative to non-alcohol related cirrhosis (FDR < 0.05). In vitro bacterial culture studies showed that physiological concentrations of ethanol and its oxidative metabolites inhibited E. coli growth in a pH- and concentration-dependent manner. Characterization of the mucosally associated gut microbiome via rectal swab revealed findings consistent with amino acid/nitrogen abundance versus carbohydrate limitation in the mucosal microenvironment as well as unique features of alcohol-related cirrhosis possibly consistent with the influence of host-derived metabolites on the composition of mucosally adherent microbiota.


Assuntos
Bactérias/isolamento & purificação , Aderência Bacteriana , Microbioma Gastrointestinal , Cirrose Hepática Alcoólica/microbiologia , Reto/microbiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Bactérias/classificação , Bactérias/genética , Fenômenos Fisiológicos Bacterianos , Estudos Transversais , Feminino , Humanos , Mucosa Intestinal/microbiologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
7.
Hepatology ; 74(6): 3427-3440, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34233020

RESUMO

BACKGROUND AND AIMS: Although germ-free mice are an indispensable tool in studying the gut microbiome and its effects on host physiology, they are phenotypically different than their conventional counterparts. While antibiotic-mediated microbiota depletion in conventional mice leads to physiologic alterations that often mimic the germ-free state, the degree to which the effects of microbial colonization on the host are reversible is unclear. The gut microbiota produce abundant short chain fatty acids (SCFAs), and previous studies have demonstrated a link between microbial-derived SCFAs and global hepatic histone acetylation in germ-free mice. APPROACH AND RESULTS: We demonstrate that global hepatic histone acetylation states measured by mass spectrometry remained largely unchanged despite loss of luminal and portal vein SCFAs after antibiotic-mediated microbiota depletion. In contrast to stable hepatic histone acetylation states, we see robust hepatic transcriptomic alterations after microbiota depletion. Additionally, neither dietary supplementation with supraphysiologic levels of SCFA nor the induction of hepatocyte proliferation in the absence of microbiota-derived SCFAs led to alterations in global hepatic histone acetylation. CONCLUSIONS: These results suggest that microbiota-dependent landscaping of the hepatic epigenome through global histone acetylation is static in nature, while the hepatic transcriptome is responsive to alterations in the gut microbiota.


Assuntos
Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal/fisiologia , Histona Acetiltransferases/metabolismo , Animais , Linhagem Celular , Masculino , Camundongos Endogâmicos C57BL
8.
Cell Host Microbe ; 29(3): 394-407.e5, 2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33440171

RESUMO

Gut microbiota metabolites may be important for host health, yet few studies investigate the correlation between human gut microbiome and production of fecal metabolites and their impact on the plasma metabolome. Since gut microbiota metabolites are influenced by diet, we performed a longitudinal analysis of the impact of three divergent diets, vegan, omnivore, and a synthetic enteral nutrition (EEN) diet lacking fiber, on the human gut microbiome and its metabolome, including after a microbiota depletion intervention. Omnivore and vegan, but not EEN, diets altered fecal amino acid levels by supporting the growth of Firmicutes capable of amino acid metabolism. This correlated with relative abundance of a sizable number of fecal amino acid metabolites, some not previously associated with the gut microbiota. The effect on the plasma metabolome, in contrast, were modest. The impact of diet, particularly fiber, on the human microbiome influences broad classes of metabolites that may modify health.


Assuntos
Fibras na Dieta , Microbioma Gastrointestinal/fisiologia , Aminoácidos , Bactérias/metabolismo , Dieta , Nutrição Enteral , Fezes/microbiologia , Firmicutes/metabolismo , Microbioma Gastrointestinal/genética , Humanos , Metaboloma , Veganos
9.
J Clin Invest ; 131(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33141762

RESUMO

As the interface between the gut microbiota and the mucosal immune system, there has been great interest in the maintenance of colonic epithelial integrity through mitochondrial oxidation of butyrate, a short-chain fatty acid produced by the gut microbiota. Herein, we showed that the intestinal epithelium could also oxidize long-chain fatty acids, and that luminally delivered acylcarnitines in bile could be consumed via apical absorption by the intestinal epithelium, resulting in mitochondrial oxidation. Finally, intestinal inflammation led to mitochondrial dysfunction in the apical domain of the surface epithelium that may reduce the consumption of fatty acids, contributing to higher concentrations of fecal acylcarnitines in murine Citrobacter rodentium-induced colitis and human inflammatory bowel disease. These results emphasized the importance of both the gut microbiota and the liver in the delivery of energy substrates for mitochondrial metabolism by the intestinal epithelium.


Assuntos
Carnitina/análogos & derivados , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/imunologia , Fígado/imunologia , Mitocôndrias/imunologia , Animais , Células CACO-2 , Carnitina/imunologia , Infecções por Enterobacteriaceae/patologia , Feminino , Humanos , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mitocôndrias/patologia
10.
EMBO Rep ; 20(6)2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31061170

RESUMO

RNA binding proteins, including IMP1/IGF2BP1, are essential regulators of intestinal development and cancer. Imp1 hypomorphic mice exhibit gastrointestinal growth defects, yet the specific role for IMP1 in colon epithelial repair is unclear. Our prior work revealed that intestinal epithelial cell-specific Imp1 deletion (Imp1ΔIEC ) was associated with better regeneration in mice after irradiation. Here, we report increased IMP1 expression in patients with Crohn's disease and ulcerative colitis. We demonstrate that Imp1ΔIEC mice exhibit enhanced recovery following dextran sodium sulfate (DSS)-mediated colonic injury. Imp1ΔIEC mice exhibit Paneth cell granule changes, increased autophagy flux, and upregulation of Atg5. In silico and biochemical analyses revealed direct binding of IMP1 to MAP1LC3B, ATG3, and ATG5 transcripts. Genetic deletion of essential autophagy gene Atg7 in Imp1ΔIEC mice revealed increased sensitivity of double-mutant mice to colonic injury compared to control or Atg7 single mutant mice, suggesting a compensatory relationship between Imp1 and the autophagy pathway. The present study defines a novel interplay between IMP1 and autophagy, where IMP1 may be transiently induced during damage to modulate colonic epithelial cell responses to damage.


Assuntos
Mucosa Intestinal/metabolismo , Proteínas de Ligação a RNA/genética , Cicatrização/genética , Adulto , Idoso , Animais , Autofagia/genética , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Biomarcadores , Estudos de Casos e Controles , Linhagem Celular , Colite Ulcerativa/genética , Colite Ulcerativa/metabolismo , Colite Ulcerativa/patologia , Colo , Doença de Crohn/genética , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Modelos Animais de Doenças , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Imuno-Histoquímica , Mucosa Intestinal/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Celulas de Paneth/metabolismo , Celulas de Paneth/patologia , Ligação Proteica , Biossíntese de Proteínas , Processamento Pós-Transcricional do RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Adulto Jovem
11.
Gastroenterology ; 155(6): 1741-1752.e5, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30144429

RESUMO

BACKGROUND & AIMS: Intestinal bacteria can modify the composition of bile acids and bile acids, which are regulated by the farnesoid X receptor, affect the survival and growth of gut bacteria. We studied the effects of obeticholic acid (OCA), a bile acid analogue and farnesoid X receptor agonist, on the intestinal microbiomes of humans and mice. METHODS: We performed a phase I study in 24 healthy volunteers given OCA (5, 10, or 25 mg/d for 17 days). Fecal and plasma specimens were collected at baseline (day 0) and on days 17 (end of dosing) and 37 (end of study). The fecal specimens were analyzed by shotgun meta-genomic sequencing. A Uniref90 high-stringency genomic analysis was used to assign specific genes to the taxonomic signature of bacteria whose abundance was associated with OCA. Male C57BL/6 mice were gavage fed daily with water, vehicle, or OCA (10 mg/kg) for 2 weeks. Small intestine luminal contents were collected by flushing with saline and fecal pellets were collected at baseline and day 14. Mouse samples were analyzed by 16S-tagged sequencing. Culture experiments were performed to determine the taxonomic-specific effects of bile acids and OCA on bacterial growth. RESULTS: Suppression of endogenous bile acid synthesis by OCA in subjects led to a reversible induction of gram-positive bacteria that are found in the small intestine and are components of the diet and oral microbiota. We found that bile acids decreased proliferation of these bacteria in minimum inhibitory concentration assays. In these organisms, there was an increase in the representation of microbial genomic pathways involved in DNA synthesis and amino acid metabolism with OCA treatment of subjects. Consistent with these findings, mice fed OCA had lower endogenous bile acid levels and an increased proportion of Firmicutes, specifically in the small intestine, compared with mice fed water or vehicle. CONCLUSIONS: In studying the effects of OCA in humans and mice, we found evidence for interactions between bile acids and features of the small intestinal microbiome. These findings indicate that farnesoid X receptor activation alters the intestinal microbiota and could provide opportunities for microbiome biomarker discovery or new approaches to engineering the human microbiome. ClinicalTrials.gov, NCT01933503.


Assuntos
Ácidos e Sais Biliares/fisiologia , Ácido Quenodesoxicólico/análogos & derivados , Microbioma Gastrointestinal/efeitos dos fármacos , Intestino Delgado/microbiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Adulto , Animais , Ácido Quenodesoxicólico/farmacocinética , Feminino , Voluntários Saudáveis , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
12.
Proc Natl Acad Sci U S A ; 115(16): 4170-4175, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610310

RESUMO

The succession from aerobic and facultative anaerobic bacteria to obligate anaerobes in the infant gut along with the differences between the compositions of the mucosally adherent vs. luminal microbiota suggests that the gut microbes consume oxygen, which diffuses into the lumen from the intestinal tissue, maintaining the lumen in a deeply anaerobic state. Remarkably, measurements of luminal oxygen levels show nearly identical pO2 (partial pressure of oxygen) profiles in conventional and germ-free mice, pointing to the existence of oxygen consumption mechanisms other than microbial respiration. In vitro experiments confirmed that the luminal contents of germ-free mice are able to chemically consume oxygen (e.g., via lipid oxidation reactions), although at rates significantly lower than those observed in the case of conventionally housed mice. For conventional mice, we also show that the taxonomic composition of the gut microbiota adherent to the gut mucosa and in the lumen throughout the length of the gut correlates with oxygen levels. At the same time, an increase in the biomass of the gut microbiota provides an explanation for the reduction of luminal oxygen in the distal vs. proximal gut. These results demonstrate how oxygen from the mammalian host is used by the gut microbiota, while both the microbes and the oxidative chemical reactions regulate luminal oxygen levels, shaping the composition of the microbial community throughout different regions of the gut.


Assuntos
Anaerobiose , Bactérias Anaeróbias/metabolismo , Microbioma Gastrointestinal , Mucosa Intestinal/metabolismo , Oxigênio/metabolismo , Animais , Bactérias Anaeróbias/isolamento & purificação , Sistemas Computacionais , Mucosa Gástrica/metabolismo , Conteúdo Gastrointestinal/química , Vida Livre de Germes , Lipídeos/química , Medições Luminescentes , Metaloporfirinas/análise , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Oxigênio/análise , Consumo de Oxigênio , Proteínas/química
13.
Sci Transl Med ; 9(416)2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29141885

RESUMO

Gut dysbiosis during inflammatory bowel disease involves alterations in the gut microbiota associated with inflammation of the host gut. We used a combination of shotgun metagenomic sequencing and metabolomics to analyze fecal samples from pediatric patients with Crohn's disease and found an association between disease severity, gut dysbiosis, and bacterial production of free amino acids. Nitrogen flux studies using 15N in mice showed that activity of bacterial urease, an enzyme that releases ammonia by hydrolysis of host urea, led to the transfer of murine host-derived nitrogen to the gut microbiota where it was used for amino acid synthesis. Inoculation of a conventional murine host (pretreated with antibiotics and polyethylene glycol) with commensal Escherichia coli engineered to express urease led to dysbiosis of the gut microbiota, resulting in a predominance of Proteobacteria species. This was associated with a worsening of immune-mediated colitis in these animals. A potential role for altered urease expression and nitrogen flux in the development of gut dysbiosis suggests that bacterial urease may be a potential therapeutic target for inflammatory bowel diseases.


Assuntos
Proteínas de Bactérias/metabolismo , Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Disbiose/metabolismo , Disbiose/microbiologia , Microbioma Gastrointestinal/fisiologia , Urease/metabolismo , Animais , Humanos , Camundongos
14.
J Clin Invest ; 125(7): 2841-50, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26098218

RESUMO

Increasing evidence indicates that the gut microbiota can be altered to ameliorate or prevent disease states, and engineering the gut microbiota to therapeutically modulate host metabolism is an emerging goal of microbiome research. In the intestine, bacterial urease converts host-derived urea to ammonia and carbon dioxide, contributing to hyperammonemia-associated neurotoxicity and encephalopathy in patients with liver disease. Here, we engineered murine gut microbiota to reduce urease activity. Animals were depleted of their preexisting gut microbiota and then inoculated with altered Schaedler flora (ASF), a defined consortium of 8 bacteria with minimal urease gene content. This protocol resulted in establishment of a persistent new community that promoted a long-term reduction in fecal urease activity and ammonia production. Moreover, in a murine model of hepatic injury, ASF transplantation was associated with decreased morbidity and mortality. These results provide proof of concept that inoculation of a prepared host with a defined gut microbiota can lead to durable metabolic changes with therapeutic utility.


Assuntos
Terapia Biológica/métodos , Sistema Digestório/microbiologia , Hiperamonemia/microbiologia , Hiperamonemia/terapia , Microbiota , Amônia/metabolismo , Animais , Bactérias/enzimologia , Bactérias/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bioengenharia , Doença Hepática Induzida por Substâncias e Drogas/terapia , Sistema Digestório/metabolismo , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Genes Bacterianos , Hiperamonemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Microbiota/fisiologia , Fatores de Tempo , Urease/genética , Urease/metabolismo
15.
Immunity ; 38(6): 1211-22, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23791643

RESUMO

The intracellular signaling molecule TRAF6 is critical for Toll-like receptor (TLR)-mediated activation of dendritic cells (DCs). We now report that DC-specific deletion of TRAF6 (TRAF6ΔDC) resulted, unexpectedly, in loss of mucosal tolerance, characterized by spontaneous development of T helper 2 (Th2) cells in the lamina propria and eosinophilic enteritis and fibrosis in the small intestine. Loss of tolerance required the presence of gut commensal microbiota but was independent of DC-expressed MyD88. Further, TRAF6ΔDC mice exhibited decreased regulatory T (Treg) cell numbers in the small intestine and diminished induction of iTreg cells in response to model antigen. Evidence suggested that this defect was associated with diminished DC expression of interleukin-2 (IL-2). Finally, we demonstrate that aberrant Th2 cell-associated responses in TRAF6ΔDC mice could be mitigated via restoration of Treg cell activity. Collectively, our findings reveal a role for TRAF6 in directing DC maintenance of intestinal immune tolerance through balanced induction of Treg versus Th2 cell immunity.


Assuntos
Células Dendríticas/imunologia , Enterite/imunologia , Eosinofilia/imunologia , Eosinófilos/imunologia , Gastrite/imunologia , Intestinos/imunologia , Linfócitos T Reguladores/imunologia , Fator 6 Associado a Receptor de TNF/metabolismo , Células Th2/imunologia , Animais , Células Cultivadas , Células Dendríticas/microbiologia , Enterite/genética , Eosinofilia/genética , Gastrite/genética , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Tolerância Imunológica/genética , Interleucina-2/genética , Interleucina-2/metabolismo , Intestinos/microbiologia , Intestinos/patologia , Ativação Linfocitária/genética , Metagenoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais/genética , Linfócitos T Reguladores/microbiologia , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/imunologia , Células Th2/microbiologia
16.
Clin Vaccine Immunol ; 18(5): 707-16, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21450977

RESUMO

We evaluated the immunogenicity and protective efficacy of a DNA vaccine expressing codon-optimized envelope glycoprotein genes of Venezuelan equine encephalitis virus (VEEV) when delivered by intramuscular electroporation. Mice vaccinated with the DNA vaccine developed robust VEEV-neutralizing antibody responses that were comparable to those observed after administration of the live-attenuated VEEV vaccine TC-83 and were completely protected from a lethal aerosol VEEV challenge. The DNA vaccine also elicited strong neutralizing antibody responses in rabbits that persisted at high levels for at least 6 months and could be boosted by a single additional electroporation administration of the DNA performed approximately 6 months after the initial vaccinations. Cynomolgus macaques that received the vaccine by intramuscular electroporation developed substantial neutralizing antibody responses and after an aerosol challenge had no detectable serum viremia and had reduced febrile reactions, lymphopenia, and clinical signs of disease compared to those of negative-control macaques. Taken together, our results demonstrate that this DNA vaccine provides a potent means of protecting against VEEV infections and represents an attractive candidate for further development.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vírus da Encefalite Equina Venezuelana/imunologia , Encefalomielite Equina Venezuelana/prevenção & controle , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Animais , Modelos Animais de Doenças , Eletroporação , Vírus da Encefalite Equina Venezuelana/genética , Encefalomielite Equina Venezuelana/patologia , Feminino , Febre/prevenção & controle , Glicoproteínas/genética , Glicoproteínas/imunologia , Linfopenia/prevenção & controle , Macaca , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Fatores de Tempo , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Viremia/prevenção & controle
17.
Birth Defects Res A Clin Mol Teratol ; 79(8): 581-94, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17676596

RESUMO

BACKGROUND: Nitric oxide (NO), produced by the nitric oxide synthase family of enzymes, mediates multiple signaling functions, and when unchecked, NO causes pathological damage. Exposure of embryos to a variety of teratogens, including carbon monoxide (CO), has been shown to increase reactive intermediates, such as NO, and recent work showed that either the excess or absence of NO caused morphological defects. While endogenous NO is known to regulate many adult tissues, its role during embryonic organogenesis and/or in mediating responses to teratogen exposure has not been explored. METHODS: We have examined here the presence of NO during normal chick embryonic organogenesis, and investigated the teratogenicity of NO through the application of sodium nitroprusside (SNP), which mimics NO overproduction, and NG-monomethyl-L-arginine (L-NMMA), which inhibits endogenous NOS activity. RESULTS: Topical treatment with SNP or L-NMMA for 18 h resulted in morphological defects, specifically in the neural tube and somites, which corresponded to sites of altered apoptosis. The location of NO was histochemically correlated with the observed morphological defects. Coadministration of SNP or L-NMMA with CO showed functional coregulation and interaction between NO and CO in chick embryonic development. CONCLUSIONS: Our results showed that regulation of NO is essential for normal axial development, that sites of altered NO expression correlate to those of altered apoptosis and dysmorphogenesis, and that CO coadministration resulted in a rectification of normal NO expression. Collectively, these results suggest that alteration in endogenous NO/CO signaling is responsible, at least in part, for the observed NO-induced teratogenesis.


Assuntos
Desenvolvimento Embrionário/efeitos dos fármacos , Defeitos do Tubo Neural/induzido quimicamente , Óxido Nítrico/metabolismo , Organogênese/fisiologia , Animais , Apoptose/efeitos dos fármacos , Monóxido de Carbono/toxicidade , Embrião de Galinha , Combinação de Medicamentos , Perda do Embrião/induzido quimicamente , Perda do Embrião/patologia , Desenvolvimento Embrionário/fisiologia , Inibidores Enzimáticos/toxicidade , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/patologia , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/metabolismo , Nitroprussiato/toxicidade , Organogênese/efeitos dos fármacos , Somitos/efeitos dos fármacos , Somitos/metabolismo , Somitos/patologia , Teratogênicos/toxicidade , ômega-N-Metilarginina/toxicidade
18.
Mol Ther ; 9(4): 607-16, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15093191

RESUMO

Gene delivery is an essential research tool for elucidating gene structure, regulation, and function in biomedical research and is the technological basis for gene therapy. However, the application of nonviral vectors in mammalian cell transfection and gene therapy is limited in that current methods require large amounts of exogenous DNA and/or exhibit high cytotoxicity and low transfection efficiency in primary cells. Here we describe the development of a novel, noninvasive gene delivery protocol using plasmid DNA vectors, based on the principle of electric field-induced molecular vibration. This method enables foreign DNA molecules to penetrate the plasma membrane and enter the cytoplasm of both primary mesenchymal progenitor cells and established cell lines of various species, at high efficiency and with low cell mortality. This procedure requires no special reagents, allows stable expression of transduced DNA, and does not interfere with the normal cellular differentiation activities of human and chick mesenchymal progenitors.


Assuntos
Campos Eletromagnéticos , Eletroporação/instrumentação , Eletroporação/métodos , Vetores Genéticos/genética , Transfecção/métodos , Adipócitos/metabolismo , Animais , Bovinos , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular , Embrião de Galinha , Condrócitos/metabolismo , Citoplasma/metabolismo , DNA/química , DNA/metabolismo , Citometria de Fluxo , Técnicas de Transferência de Genes , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C3H , Microscopia de Fluorescência , Plasmídeos/metabolismo , Células-Tronco/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...