Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38895199

RESUMO

Dose-limiting toxicities remain a major barrier to drug development and therapy, revealing the limited predictive power of human genetics. Herein, we demonstrate the utility of a more comprehensive approach to studying drug toxicity through longitudinal study of the human gut microbiome during colorectal cancer (CRC) treatment (NCT04054908) coupled to cell culture and mouse experiments. 16S rRNA gene and metagenomic sequencing revealed significant shifts in gut microbial community structure during treatment with oral fluoropyrimidines, which was validated in an independent cohort. Gene abundance was also markedly changed by oral fluoropyrimidines, including an enrichment for the preTA operon, which is sufficient for the inactivation of active metabolite 5-fluorouracil (5-FU). Higher levels of preTA led to increased 5-FU depletion by the gut microbiota grown ex vivo. Germ-free and antibiotic-treated mice had increased fluoropyrimidine toxicity, which was rescued by colonization with the mouse gut microbiota, preTA+ E. coli, or CRC patient stool with high preTA levels. preTA abundance was negatively associated with patient toxicities. Together, these data support a causal, clinically relevant interaction between a human gut bacterial operon and the dose-limiting side effects of cancer treatment. Our approach is generalizable to other drugs, including cancer immunotherapies, and provides valuable insights into host-microbiome interactions in the context of disease.

2.
Cell Rep ; 42(12): 113521, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38070135

RESUMO

The gut microbiome modulates seizure susceptibility and the anti-seizure effects of the ketogenic diet (KD) in animal models, but whether these relationships translate to KD therapies for human epilepsy is unclear. We find that the clinical KD alters gut microbial function in children with refractory epilepsy. Colonizing mice with KD-associated microbes promotes seizure resistance relative to matched pre-treatment controls. Select metagenomic and metabolomic features, including those related to anaplerosis, fatty acid ß-oxidation, and amino acid metabolism, are seen with human KD therapy and preserved upon microbiome transfer to mice. Mice colonized with KD-associated gut microbes exhibit altered hippocampal transcriptomes, including pathways related to ATP synthesis, glutathione metabolism, and oxidative phosphorylation, and are linked to susceptibility genes identified in human epilepsy. Our findings reveal key microbial functions that are altered by KD therapies for pediatric epilepsy and linked to microbiome-induced alterations in brain gene expression and seizure protection in mice.


Assuntos
Dieta Cetogênica , Epilepsia , Microbioma Gastrointestinal , Microbiota , Criança , Humanos , Animais , Camundongos , Corpos Cetônicos , Modelos Animais de Doenças , Convulsões
3.
mSystems ; 7(5): e0099521, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36047699

RESUMO

Microbial source tracking analysis has emerged as a widespread technique for characterizing the properties of complex microbial communities. However, this analysis is currently limited to source environments sampled in a specific study. In order to expand the scope beyond one single study and allow the exploration of source environments using large databases and repositories, such as the Earth Microbiome Project, a source selection procedure is required. Such a procedure will allow differentiating between contributing environments and nuisance ones when the number of potential sources considered is high. Here, we introduce STENSL (microbial Source Tracking with ENvironment SeLection), a machine learning method that extends common microbial source tracking analysis by performing an unsupervised source selection and enabling sparse identification of latent source environments. By incorporating sparsity into the estimation of potential source environments, STENSL improves the accuracy of true source contribution, while significantly reducing the noise introduced by noncontributing ones. We therefore anticipate that source selection will augment microbial source tracking analyses, enabling exploration of multiple source environments from publicly available repositories while maintaining high accuracy of the statistical inference. IMPORTANCE Microbial source tracking is a powerful tool to characterize the properties of complex microbial communities. However, this analysis is currently limited to source environments sampled in a specific study. In many applications there is a clear need to consider source selection over a large array of microbial environments, external to the study. To this end, we developed STENSL (microbial Source Tracking with ENvironment SeLection), an expectation-maximization algorithm with sparsity that enables the identification of contributing sources among a large set of potential microbial environments. With the unprecedented expansion of microbiome data repositories such as the Earth Microbiome Project, recording over 200,000 samples from more than 50 types of categorized environments, STENSL takes the first steps in performing automated source exploration and selection. STENSL is significantly more accurate in identifying the contributing sources as well as the unknown source, even when considering hundreds of potential source environments, settings in which state-of-the-art microbial source tracking methods add considerable error.


Assuntos
Microbiota , Algoritmos , Aprendizado de Máquina
4.
Nat Microbiol ; 7(6): 745-746, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35606423
5.
Cell Host Microbe ; 29(9): 1378-1392.e6, 2021 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-34358434

RESUMO

Many genetic and environmental factors increase susceptibility to cognitive impairment (CI), and the gut microbiome is increasingly implicated. However, the identity of gut microbes associated with CI risk, their effects on CI, and their mechanisms remain unclear. Here, we show that a carbohydrate-restricted (ketogenic) diet potentiates CI induced by intermittent hypoxia in mice and alters the gut microbiota. Depleting the microbiome reduces CI, whereas transplantation of the risk-associated microbiome or monocolonization with Bilophila wadsworthia confers CI in mice fed a standard diet. B. wadsworthia and the risk-associated microbiome disrupt hippocampal synaptic plasticity, neurogenesis, and gene expression. The CI is associated with microbiome-dependent increases in intestinal interferon-gamma (IFNg)-producing Th1 cells. Inhibiting Th1 cell development abrogates the adverse effects of both B. wadsworthia and environmental risk factors on CI. Together, these findings identify select gut bacteria that contribute to environmental risk for CI in mice by promoting inflammation and hippocampal dysfunction.


Assuntos
Bilophila/metabolismo , Disfunção Cognitiva/patologia , Dieta Cetogênica/efeitos adversos , Hipocampo/fisiopatologia , Hipóxia Encefálica/fisiopatologia , Células Th1/imunologia , Animais , Microbioma Gastrointestinal/fisiologia , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Th1/citologia
6.
Transl Psychiatry ; 11(1): 194, 2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33790226

RESUMO

Emerging evidence highlights a critical relationship between gut microbiota and neurocognitive development. Excessive consumption of sugar and other unhealthy dietary factors during early life developmental periods yields changes in the gut microbiome as well as neurocognitive impairments. However, it is unclear whether these two outcomes are functionally connected. Here we explore whether excessive early life consumption of added sugars negatively impacts memory function via the gut microbiome. Rats were given free access to a sugar-sweetened beverage (SSB) during the adolescent stage of development. Memory function and anxiety-like behavior were assessed during adulthood and gut bacterial and brain transcriptome analyses were conducted. Taxa-specific microbial enrichment experiments examined the functional relationship between sugar-induced microbiome changes and neurocognitive and brain transcriptome outcomes. Chronic early life sugar consumption impaired adult hippocampal-dependent memory function without affecting body weight or anxiety-like behavior. Adolescent SSB consumption during adolescence also altered the gut microbiome, including elevated abundance of two species in the genus Parabacteroides (P. distasonis and P. johnsonii) that were negatively correlated with hippocampal function. Transferred enrichment of these specific bacterial taxa in adolescent rats impaired hippocampal-dependent memory during adulthood. Hippocampus transcriptome analyses revealed that early life sugar consumption altered gene expression in intracellular kinase and synaptic neurotransmitter signaling pathways, whereas Parabacteroides microbial enrichment altered gene expression in pathways associated with metabolic function, neurodegenerative disease, and dopaminergic signaling. Collectively these results identify a role for microbiota "dysbiosis" in mediating the detrimental effects of early life unhealthy dietary factors on hippocampal-dependent memory function.


Assuntos
Microbioma Gastrointestinal , Doenças Neurodegenerativas , Animais , Açúcares da Dieta/efeitos adversos , Memória , Ratos , Ratos Sprague-Dawley
7.
J Nutr ; 150(10): 2716-2728, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-32856048

RESUMO

BACKGROUND: It is unclear how high fructose consumption induces disparate metabolic responses in genetically diverse mouse strains. OBJECTIVE: We aimed to investigate whether the gut microbiota contributes to differential metabolic responses to fructose. METHODS: Eight-week-old male C57BL/6J (B6), DBA/2J (DBA), and FVB/NJ (FVB) mice were given 8% fructose solution or regular water (control) for 12 wk. The gut microbiota composition in cecum and feces was analyzed using 16S ribosomal DNA sequencing, and permutational multivariate ANOVA (PERMANOVA) was used to compare community across mouse strains, treatments, and time points. Microbiota abundance was correlated with metabolic phenotypes and host gene expression in hypothalamus, liver, and adipose tissues using Biweight midcorrelation. To test the causal role of the gut microbiota in determining fructose response, we conducted fecal transplants from B6 to DBA mice and vice versa for 4 wk, as well as gavaged antibiotic-treated DBA mice with Akkermansia for 9 wk, accompanied with or without fructose treatment. RESULTS: Compared with B6 and FVB, DBA mice had significantly higher Firmicutes to Bacteroidetes ratio and lower baseline abundance of Akkermansia and S24-7 (P < 0.05), accompanied by metabolic dysregulation after fructose consumption. Fructose altered specific microbial taxa in individual mouse strains, such as a 7.27-fold increase in Akkermansia in B6 and 0.374-fold change in Rikenellaceae in DBA (false discovery rate <5%), which demonstrated strain-specific correlations with host metabolic and transcriptomic phenotypes. Fecal transplant experiments indicated that B6 microbes conferred resistance to fructose-induced weight gain in DBA mice (F = 43.1, P < 0.001), and Akkermansia colonization abrogated the fructose-induced weight gain (F = 17.8, P < 0.001) and glycemic dysfunctions (F = 11.8, P = 0.004) in DBA mice. CONCLUSIONS: Our findings support that differential microbiota composition between mouse strains is partially responsible for host metabolic sensitivity to fructose, and that Akkermansia is a key bacterium that confers resistance to fructose-induced metabolic dysregulation.


Assuntos
Bactérias/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Frutose/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Animais , Ceco/microbiologia , Transplante de Microbiota Fecal , Fezes/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos , Distribuição Aleatória
8.
Cell Metab ; 31(6): 1049-1051, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32492391

RESUMO

The ketogenic diet is used to treat neurological and metabolic symptoms of disease, but the extent of its influences across organ systems remains unclear. Ang et al., 2020 reveal that ketone bodies induced by the diet inhibit specific bacteria of the gut microbiota and suppress pro-inflammatory T cells in the intestine.


Assuntos
Dieta Cetogênica , Microbioma Gastrointestinal , Bifidobacterium , Ésteres , Intestinos , Corpos Cetônicos , Células Th17
9.
Neurobiol Dis ; 135: 104576, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31445165

RESUMO

The gut microbiome is emerging as a key regulator of brain function and behavior and is associated with symptoms of several neurological disorders. There is emerging evidence that alterations in the gut microbiota are seen in epilepsy and in response to seizure interventions. In this review, we highlight recent studies reporting that individuals with refractory epilepsy exhibit altered composition of the gut microbiota. We further discuss antibiotic treatment and infection as microbiome-related factors that influence seizure susceptibility in humans and animal models. In addition, we evaluate how the microbiome may mediate effects of the ketogenic diet, probiotic treatment, and anti-epileptic drugs on reducing both seizure frequency and severity. Finally, we assess the open questions in interrogating roles for the microbiome in epilepsy and address the prospect that continued research may uncover fundamental insights for understanding risk factors for epilepsy, as well as novel approaches for treating refractory epilepsy.


Assuntos
Epilepsia/etiologia , Microbioma Gastrointestinal/fisiologia , Papel (figurativo) , Convulsões/complicações , Animais , Dieta Cetogênica , Epilepsia Resistente a Medicamentos/etiologia , Epilepsia Resistente a Medicamentos/microbiologia , Epilepsia/microbiologia , Humanos
11.
Cell ; 173(7): 1728-1741.e13, 2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29804833

RESUMO

The ketogenic diet (KD) is used to treat refractory epilepsy, but the mechanisms underlying its neuroprotective effects remain unclear. Here, we show that the gut microbiota is altered by the KD and required for protection against acute electrically induced seizures and spontaneous tonic-clonic seizures in two mouse models. Mice treated with antibiotics or reared germ free are resistant to KD-mediated seizure protection. Enrichment of, and gnotobiotic co-colonization with, KD-associated Akkermansia and Parabacteroides restores seizure protection. Moreover, transplantation of the KD gut microbiota and treatment with Akkermansia and Parabacteroides each confer seizure protection to mice fed a control diet. Alterations in colonic lumenal, serum, and hippocampal metabolomic profiles correlate with seizure protection, including reductions in systemic gamma-glutamylated amino acids and elevated hippocampal GABA/glutamate levels. Bacterial cross-feeding decreases gamma-glutamyltranspeptidase activity, and inhibiting gamma-glutamylation promotes seizure protection in vivo. Overall, this study reveals that the gut microbiota modulates host metabolism and seizure susceptibility in mice.


Assuntos
Dieta Cetogênica , Microbioma Gastrointestinal , Convulsões/dietoterapia , Animais , Antibacterianos/farmacologia , Bacteroides/efeitos dos fármacos , Bacteroides/genética , Bacteroides/isolamento & purificação , Modelos Animais de Doenças , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Mucosa Intestinal/metabolismo , Canal de Potássio Kv1.1/deficiência , Canal de Potássio Kv1.1/genética , Metaboloma/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Análise de Componente Principal , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Convulsões/patologia , Ácido gama-Aminobutírico/metabolismo , gama-Glutamiltransferase/metabolismo
12.
Nat Neurosci ; 20(2): 145-155, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28092661

RESUMO

The diverse collection of microorganisms that inhabit the gastrointestinal tract, collectively called the gut microbiota, profoundly influences many aspects of host physiology, including nutrient metabolism, resistance to infection and immune system development. Studies investigating the gut-brain axis demonstrate a critical role for the gut microbiota in orchestrating brain development and behavior, and the immune system is emerging as an important regulator of these interactions. Intestinal microbes modulate the maturation and function of tissue-resident immune cells in the CNS. Microbes also influence the activation of peripheral immune cells, which regulate responses to neuroinflammation, brain injury, autoimmunity and neurogenesis. Accordingly, both the gut microbiota and immune system are implicated in the etiopathogenesis or manifestation of neurodevelopmental, psychiatric and neurodegenerative diseases, such as autism spectrum disorder, depression and Alzheimer's disease. In this review, we discuss the role of CNS-resident and peripheral immune pathways in microbiota-gut-brain communication during health and neurological disease.


Assuntos
Transtorno do Espectro Autista/imunologia , Encéfalo/imunologia , Trato Gastrointestinal/imunologia , Microbiota/imunologia , Doenças do Sistema Nervoso/imunologia , Animais , Autoimunidade/fisiologia , Humanos , Doenças do Sistema Nervoso/fisiopatologia
14.
Ann Plast Surg ; 74 Suppl 1: S15-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25774967

RESUMO

Radical oncologic resection can result in large soft tissue defects with exposure of underlying vessels. Unless immediately covered with viable soft tissue, these vessels are vulnerable to desiccation from air exposure and mechanical trauma. Local radiation treatment also contributes to a decline in vessel wall strength. We present an index case of a patient with prolonged exposure of her femoral bone and superficial femoral artery after an initial failed reconstruction of a soft tissue sarcoma resection defect. We provided coverage using a free latissimus dorsi muscle flap. Two weeks after the initial free flap operation, the patient was readmitted to emergency service with profuse bleeding from beneath the free flap. Intraoperative inspection revealed a 2-cm defect of the irradiated superficial femoral artery. The defect was repaired with cryopreserved human arterial graft, and the flap was reset. This case highlights the importance of immediate coverage of soft tissue defects after oncologic resection. If any vessels are left exposed, they should be closely inspected before a delayed flap coverage to rule out future sources of bleeding that may jeopardize the outcomes of an otherwise successful free flap operation.


Assuntos
Artéria Femoral/lesões , Artéria Femoral/efeitos da radiação , Retalhos de Tecido Biológico , Lesões por Radiação/complicações , Coxa da Perna/cirurgia , Feminino , Humanos , Pessoa de Meia-Idade , Ruptura/etiologia , Sarcoma/radioterapia , Sarcoma/cirurgia , Neoplasias de Tecidos Moles/radioterapia , Neoplasias de Tecidos Moles/cirurgia
15.
Ann Plast Surg ; 74(5): 584-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25643192

RESUMO

BACKGROUND: Schwann cell-like cells differentiated from adipose-derived stem cells may have an important role in peripheral nerve regeneration. Herein, we document the individual effects of growth factors in Schwann cell-like differentiation medium. METHODS: There were 6 groups in the study. In the control group, we supplemented the rat adipose-derived stem cells with normal cell culture medium. In group 1, we fed the cells with Schwann cell-like differentiation medium (normal cell culture medium supplemented with platelet-derived growth factor, basic fibroblast growth factor, forskolin, and glial growth factor). In the other groups, we removed the components of the medium one at a time from the differentiation medium so that group 2 lacked glial growth factor, group 3 lacked forskolin, group 4 lacked basic fibroblast growth factor, and group 5 lacked platelet-derived growth factor. We examined the expression of the Schwann cell-specific genes with quantitative reverse transcription polymerase chain reaction and immunofluorescence staining in each group. RESULTS: Groups 3 and 4, lacking forskolin and basic fibroblast growth factor, respectively, had the highest expression levels of integrin-ß4, and p75. Group 1 showed a 3.2-fold increase in the expression of S100, but the expressions of integrin-ß4 and p75 were significantly lower compared to groups 3 and 4. Group 2 [glial growth factor (-)] did not express significant levels of Schwann cell-specific genes. The gene expression profile in group 4 most closely resembled Schwann cells. Immunofluorescence staining results were parallel with the quantitative real-time polymerase chain reaction results. CONCLUSIONS: Glial growth factor is a key component of Schwann cell-like differentiation medium.


Assuntos
Tecido Adiposo/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Meios de Cultura/química , Expressão Gênica/fisiologia , Células-Tronco Mesenquimais/fisiologia , Células de Schwann/fisiologia , Animais , Técnicas de Cultura de Células/instrumentação , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Regeneração Nervosa/fisiologia , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...