Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Gut ; 72(5): 918-928, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36627187

RESUMO

OBJECTIVE: Gestational diabetes mellitus (GDM) is a condition in which women without diabetes are diagnosed with glucose intolerance during pregnancy, typically in the second or third trimester. Early diagnosis, along with a better understanding of its pathophysiology during the first trimester of pregnancy, may be effective in reducing incidence and associated short-term and long-term morbidities. DESIGN: We comprehensively profiled the gut microbiome, metabolome, inflammatory cytokines, nutrition and clinical records of 394 women during the first trimester of pregnancy, before GDM diagnosis. We then built a model that can predict GDM onset weeks before it is typically diagnosed. Further, we demonstrated the role of the microbiome in disease using faecal microbiota transplant (FMT) of first trimester samples from pregnant women across three unique cohorts. RESULTS: We found elevated levels of proinflammatory cytokines in women who later developed GDM, decreased faecal short-chain fatty acids and altered microbiome. We next confirmed that differences in GDM-associated microbial composition during the first trimester drove inflammation and insulin resistance more than 10 weeks prior to GDM diagnosis using FMT experiments. Following these observations, we used a machine learning approach to predict GDM based on first trimester clinical, microbial and inflammatory markers with high accuracy. CONCLUSION: GDM onset can be identified in the first trimester of pregnancy, earlier than currently accepted. Furthermore, the gut microbiome appears to play a role in inflammation-induced GDM pathogenesis, with interleukin-6 as a potential contributor to pathogenesis. Potential GDM markers, including microbiota, can serve as targets for early diagnostics and therapeutic intervention leading to prevention.


Assuntos
Diabetes Gestacional , Microbiota , Gravidez , Feminino , Humanos , Diabetes Gestacional/diagnóstico , Terceiro Trimestre da Gravidez , Inflamação , Citocinas
2.
Bioinformatics ; 38(6): 1615-1623, 2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-34999748

RESUMO

MOTIVATION: Recent technological developments have facilitated an expansion of microbiome-metabolome studies, in which samples are assayed using both genomic and metabolomic technologies to characterize the abundances of microbial taxa and metabolites. A common goal of these studies is to identify microbial species or genes that contribute to differences in metabolite levels across samples. Previous work indicated that integrating these datasets with reference knowledge on microbial metabolic capacities may enable more precise and confident inference of microbe-metabolite links. RESULTS: We present MIMOSA2, an R package and web application for model-based integrative analysis of microbiome-metabolome datasets. MIMOSA2 uses genomic and metabolic reference databases to construct a community metabolic model based on microbiome data and uses this model to predict differences in metabolite levels across samples. These predictions are compared with metabolomics data to identify putative microbiome-governed metabolites and taxonomic contributors to metabolite variation. MIMOSA2 supports various input data types and customization with user-defined metabolic pathways. We establish MIMOSA2's ability to identify ground truth microbial mechanisms in simulation datasets, compare its results with experimentally inferred mechanisms in honeybee microbiota, and demonstrate its application in two human studies of inflammatory bowel disease. Overall, MIMOSA2 combines reference databases, a validated statistical framework, and a user-friendly interface to facilitate modeling and evaluating relationships between members of the microbiota and their metabolic products. AVAILABILITY AND IMPLEMENTATION: MIMOSA2 is implemented in R under the GNU General Public License v3.0 and is freely available as a web server at http://elbo-spice.cs.tau.ac.il/shiny/MIMOSA2shiny/ and as an R package from http://www.borensteinlab.com/software_MIMOSA2.html. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Metaboloma , Microbiota , Animais , Humanos , Metabolômica/métodos , Microbiota/genética , Software , Redes e Vias Metabólicas
3.
BMC Med ; 18(1): 281, 2020 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-33081767

RESUMO

BACKGROUND: Adjuvant chemotherapy induces weight gain, glucose intolerance, and hypertension in about a third of women. The mechanisms underlying these events have not been defined. This study assessed the association between the microbiome and weight gain in patients treated with adjuvant chemotherapy for breast and gynecological cancers. METHODS: Patients were recruited before starting adjuvant therapy. Weight and height were measured before treatment and 4-6 weeks after treatment completion. Weight gain was defined as an increase of 3% or more in body weight. A stool sample was collected before treatment, and 16S rRNA gene sequencing was performed. Data regarding oncological therapy, menopausal status, and antibiotic use was prospectively collected. Patients were excluded if they were treated by antibiotics during the study. Fecal transplant experiments from patients were conducted using Swiss Webster germ-free mice. RESULTS: Thirty-three patients were recruited; of them, 9 gained 3.5-10.6% of baseline weight. The pretreatment microbiome of women who gained weight following treatment was significantly different in diversity and taxonomy from that of control women. Fecal microbiota transplantation from pretreatment samples of patients that gained weight induced metabolic changes in germ-free mice compared to mice transplanted with pretreatment fecal samples from the control women. CONCLUSION: The microbiome composition is predictive of weight gain following adjuvant chemotherapy and induces adverse metabolic changes in germ-free mice, suggesting it contributes to adverse metabolic changes seen in patients. Confirmation of these results in a larger patient cohort is warranted.


Assuntos
Neoplasias da Mama/complicações , Quimioterapia Adjuvante/efeitos adversos , Microbioma Gastrointestinal/genética , Neoplasias dos Genitais Femininos/complicações , Aumento de Peso/efeitos dos fármacos , Adolescente , Adulto , Idoso , Animais , Neoplasias da Mama/tratamento farmacológico , Estudos de Coortes , Feminino , Neoplasias dos Genitais Femininos/tratamento farmacológico , Humanos , Camundongos , Pessoa de Meia-Idade , Adulto Jovem
5.
bioRxiv ; 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-37461534

RESUMO

The human gut microbiome is a complex ecosystem with profound implications for health and disease. This recognition has led to a surge in multi-omic microbiome studies, employing various molecular assays to elucidate the microbiome's role in diseases across multiple functional layers. However, despite the clear value of these multi-omic datasets, rigorous integrative analysis of such data poses significant challenges, hindering a comprehensive understanding of microbiome-disease interactions. Perhaps most notably, multiple approaches, including univariate and multivariate analyses, as well as machine learning, have been applied to such data to identify disease-associated markers, namely, specific features (e.g., species, pathways, metabolites) that are significantly altered in disease state. These methods, however, often yield extensive lists of features associated with the disease without effectively capturing the multi-layered structure of multi-omic data or offering clear, interpretable hypotheses about underlying microbiome-disease mechanisms. Here, we address this challenge by introducing MintTea - an intermediate integration-based method for analyzing multi-omic microbiome data. MintTea combines a canonical correlation analysis (CCA) extension, consensus analysis, and an evaluation protocol to robustly identify disease-associated multi-omic modules. Each such module consists of a set of features from the various omics that both shift in concord, and collectively associate with the disease. Applying MintTea to diverse case-control cohorts with multi-omic data, we show that this framework is able to capture modules with high predictive power for disease, significant cross-omic correlations, and alignment with known microbiome-disease associations. For example, analyzing samples from a metabolic syndrome (MS) study, we found a MS-associated module comprising of a highly correlated cluster of serum glutamate- and TCA cycle-related metabolites, as well as bacterial species previously implicated in insulin resistance. In another cohort, we identified a module associated with late-stage colorectal cancer, featuring Peptostreptococcus and Gemella species and several fecal amino acids, in agreement with these species' reported role in the metabolism of these amino acids and their coordinated increase in abundance during disease development. Finally, comparing modules identified in different datasets, we detected multiple significant overlaps, suggesting common interactions between microbiome features. Combined, this work serves as a proof of concept for the potential benefits of advanced integration methods in generating integrated multi-omic hypotheses underlying microbiome-disease interactions and a promising avenue for researchers seeking systems-level insights into coherent mechanisms governing microbiome-related diseases.

6.
Nat Commun ; 15(1): 2621, 2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38521774

RESUMO

Multi-omic studies of the human gut microbiome are crucial for understanding its role in disease across multiple functional layers. Nevertheless, integrating and analyzing such complex datasets poses significant challenges. Most notably, current analysis methods often yield extensive lists of disease-associated features (e.g., species, pathways, or metabolites), without capturing the multi-layered structure of the data. Here, we address this challenge by introducing "MintTea", an intermediate integration-based approach combining canonical correlation analysis extensions, consensus analysis, and an evaluation protocol. MintTea identifies "disease-associated multi-omic modules", comprising features from multiple omics that shift in concord and that collectively associate with the disease. Applied to diverse cohorts, MintTea captures modules with high predictive power, significant cross-omic correlations, and alignment with known microbiome-disease associations. For example, analyzing samples from a metabolic syndrome study, MintTea identifies a module with serum glutamate- and TCA cycle-related metabolites, along with bacterial species linked to insulin resistance. In another dataset, MintTea identifies a module associated with late-stage colorectal cancer, including Peptostreptococcus and Gemella species and fecal amino acids, in line with these species' metabolic activity and their coordinated gradual increase with cancer development. This work demonstrates the potential of advanced integration methods in generating systems-level, multifaceted hypotheses underlying microbiome-disease interactions.


Assuntos
Microbioma Gastrointestinal , Microbiota , Humanos , Multiômica , Microbiota/genética , Bactérias/genética , Microbioma Gastrointestinal/genética
7.
Autism Res ; 16(6): 1225-1235, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37119025

RESUMO

Some individuals with autism spectrum disorder (ASD) demonstrate marked behavioral improvements during febrile episodes, in what is perhaps the only present-day means of modulating the core ASD phenotype. Understanding the nature of this so-called fever effect is therefore essential for leveraging this natural temporary relief of symptoms to a sustained efficacious intervention. Toward this goal, we used machine learning to analyze the rich clinical data of the Simons Simplex Collection, in which one out of every six children with ASD was reported to improve during febrile episodes, across multiple ASD domains. Reported behavioral improvements during febrile episodes were associated with maternal infection in pregnancy (OR = 1.7, 95% CI = [1.42, 2.03], P = 4.24 × 10-4 ) and gastrointestinal (GI) dysfunction (OR = 1.46, 95% CI = [1.15, 1.81], P = 1.94 × 10-3 ). Family members of children reported to improve when febrile have an increased prevalence of autoimmune disorders (OR = 1.43, 95% CI = [1.23, 1.67], P = 3.0 × 10-6 ), language disorders (OR = 1.63, 95% CI = [1.29, 2.04], P = 2.5 × 10-5 ), and neuropsychiatric disorders (OR = 1.59, 95% CI = [1.34, 1.89], P < 1 × 10-6 ). Since both GI abnormalities and maternal immune activation have been linked to ASD via proinflammatory cytokines, these results might suggest a possible involvement of immune dysregulation in the fever effect, consistent with findings in mouse models. This work advances our understanding of the fever-responsive ASD subtype and motivates the future studies to directly test the link between proinflammatory cytokines and behavioral modifications in individuals with ASD.


Assuntos
Transtorno do Espectro Autista , Gastroenteropatias , Gravidez , Feminino , Animais , Camundongos , Transtorno do Espectro Autista/complicações , Transtorno do Espectro Autista/epidemiologia , Transtorno do Espectro Autista/diagnóstico , Febre/complicações , Febre/epidemiologia , Gastroenteropatias/complicações , Fenótipo , Citocinas
8.
NPJ Biofilms Microbiomes ; 8(1): 79, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36243731

RESUMO

Integrative analysis of microbiome and metabolome data obtained from human fecal samples is a promising avenue for better understanding the interplay between bacteria and metabolites in the human gut, in both health and disease. However, acquiring, processing, and unifying such datasets from multiple sources is a daunting and challenging task. Here we present a publicly available, simple-to-use, curated dataset collection of paired fecal microbiome-metabolome data from multiple cohorts. This data resource allows researchers to easily obtain multiple fully processed and integrated microbiome-metabolome datasets, facilitating the discovery of universal microbe-metabolite links, benchmark various microbiome-metabolome integration tools, and compare newly identified microbe-metabolite findings to other published datasets.


Assuntos
Microbioma Gastrointestinal , Fezes/microbiologia , Humanos , Metaboloma , Metabolômica , RNA Ribossômico 16S
9.
Microbiome ; 9(1): 203, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34641974

RESUMO

BACKGROUND: Microbiome-metabolome studies of the human gut have been gaining popularity in recent years, mostly due to accumulating evidence of the interplay between gut microbes, metabolites, and host health. Statistical and machine learning-based methods have been widely applied to analyze such paired microbiome-metabolome data, in the hope of identifying metabolites that are governed by the composition of the microbiome. Such metabolites can be likely modulated by microbiome-based interventions, offering a route for promoting gut metabolic health. Yet, to date, it remains unclear whether findings of microbially associated metabolites in any single study carry over to other studies or cohorts, and how robust and universal are microbiome-metabolites links. RESULTS: In this study, we addressed this challenge by performing a comprehensive meta-analysis to identify human gut metabolites that can be predicted based on the composition of the gut microbiome across multiple studies. We term such metabolites "robustly well-predicted". To this end, we processed data from 1733 samples from 10 independent human gut microbiome-metabolome studies, focusing initially on healthy subjects, and implemented a machine learning pipeline to predict metabolite levels in each dataset based on the composition of the microbiome. Comparing the predictability of each metabolite across datasets, we found 97 robustly well-predicted metabolites. These include metabolites involved in important microbial pathways such as bile acid transformations and polyamines metabolism. Importantly, however, other metabolites exhibited large variation in predictability across datasets, suggesting a cohort- or study-specific relationship between the microbiome and the metabolite. Comparing taxonomic contributors to different models, we found that some robustly well-predicted metabolites were predicted by markedly different sets of taxa across datasets, suggesting that some microbially associated metabolites may be governed by different members of the microbiome in different cohorts. We finally examined whether models trained on a control group of a given study successfully predicted the metabolite's level in the disease group of the same study, identifying several metabolites where the model was not transferable, indicating a shift in microbial metabolism in disease-associated dysbiosis. CONCLUSIONS: Combined, our findings provide a better understanding of the link between the microbiome and metabolites and allow researchers to put identified microbially associated metabolites within the context of other studies. Video abstract.


Assuntos
Microbioma Gastrointestinal , Microbiota , Ácidos e Sais Biliares , Disbiose , Microbioma Gastrointestinal/genética , Humanos , Metaboloma , Metabolômica
10.
Genome Med ; 12(1): 92, 2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-33109272

RESUMO

BACKGROUND: Multiple studies suggest a key role for gut microbiota in IgE-mediated food allergy (FA) development, but to date, none has studied it in the persistent state. METHODS: To characterize the gut microbiota composition and short-chain fatty acid (SCFAs) profiles associated with major food allergy groups, we recruited 233 patients with FA including milk (N = 66), sesame (N = 38), peanut (N = 71), and tree nuts (N = 58), and non-allergic controls (N = 58). DNA was isolated from fecal samples, and 16S rRNA gene sequences were analyzed. SCFAs in stool were analyzed from patients with a single allergy (N = 84) and controls (N = 31). RESULTS: The gut microbiota composition of allergic patients was significantly different compared to age-matched controls both in α-diversity and ß-diversity. Distinct microbial signatures were noted for FA to different foods. Prevotella copri (P. copri) was the most overrepresented species in non-allergic controls. SCFAs levels were significantly higher in the non-allergic compared to the FA groups, whereas P. copri significantly correlated with all three SCFAs. We used these microbial differences to distinguish between FA patients and non-allergic healthy controls with an area under the curve of 0.90, and for the classification of FA patients according to their FA types using a supervised learning algorithm. Bacteroides and P. copri were identified as taxa potentially contributing to KEGG acetate-related pathways enriched in non-allergic compared to FA. In addition, overall pathway dissimilarities were found among different FAs. CONCLUSIONS: Our results demonstrate a link between IgE-mediated FA and the composition and metabolic activity of the gut microbiota.


Assuntos
Suscetibilidade a Doenças , Hipersensibilidade Alimentar/etiologia , Imunoglobulina E/imunologia , Microbiota , Idoso , Idoso de 80 Anos ou mais , Biomarcadores , Ácidos Graxos Voláteis/metabolismo , Feminino , Hipersensibilidade Alimentar/metabolismo , Microbioma Gastrointestinal , Humanos , Aprendizado de Máquina , Masculino , Microbiota/imunologia , Pessoa de Meia-Idade , Probióticos , RNA Ribossômico 16S/genética
11.
Blood Adv ; 4(13): 2912-2917, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32598476

RESUMO

Oral mucositis (OM) is a common debilitating dose-limiting toxicity of cancer treatment, including hematopoietic stem cell transplantation (HSCT). We hypothesized that the oral microbiome is disturbed during allogeneic HSCT, partially accounting for the variability in OM severity. Using 16S ribosomal RNA gene sequence analysis, metabolomic profiling, and computational methods, we characterized the behavior of the salivary microbiome and metabolome of 184 patients pre- and post-HSCT. Transplantation was associated with a decrease in oral α diversity in all patients. In contrast to the gut microbiome, an association with overall survival was not detected. Among 135 patients given methotrexate for graft-versus-host disease prophylaxis pre-HSCT, Kingella and Atopobium abundance correlated with future development of severe OM. Posttransplant, Methylobacterium species were significantly enriched in patients with severe OM. Moreover, the oral microbiome and metabolome of severe OM patients underwent distinct changes post-HSCT, compared with patients with no or mild OM. Changes in specific metabolites were well explained by microbial composition, and the common metabolic pathway was the polyamines pathway, which is essential for epithelial homeostasis. Together, our findings suggest that salivary microbial composition and metabolites are associated with the development of OM, offering new insights on pathophysiology and potential avenues of intervention.


Assuntos
Microbioma Gastrointestinal , Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Microbiota , Estomatite , Doença Enxerto-Hospedeiro/etiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Estomatite/etiologia
12.
JCO Clin Cancer Inform ; 2: 1-8, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30652563

RESUMO

PURPOSE: To evaluate in a sample of adults who had been noncompliant with colorectal cancer (CRC) screening whether screening could be enhanced by an automated patient recall system based on identifying high-risk individuals using the ColonFlag test and an electronic medical record database. METHODS: A total of 79,671 individuals who were determined to be noncompliant with current screening recommendations were identified in the Maccabi Health Services program in Israel. Their cancer risk was determined by ColonFlag using information on age, sex, and CBC results. Doctors of individuals who were flagged as high risk were notified and asked to follow up with their patients. RESULTS: The ColonFlag identified 688 men and women who scored in the highest 0.87 percentile. Of these individuals, 254 had colonoscopies performed by Maccabi physicians, and 19 CRCs (7.5%) were found. An additional 15 cancers primarily identified outside of Maccabi were found through code matching. CONCLUSION: The ColonFlag test is a rapid, efficient, and inexpensive test that can be applied to scan electronic medical records to identify individuals at high risk of CRC who would otherwise avoid screening.


Assuntos
Colonoscopia/métodos , Neoplasias Colorretais/diagnóstico , Diagnóstico por Computador/métodos , Detecção Precoce de Câncer/normas , Registros Eletrônicos de Saúde/estatística & dados numéricos , Idoso , Neoplasias Colorretais/etiologia , Feminino , Seguimentos , Sistemas Pré-Pagos de Saúde , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA