Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 24(12): 2108-2120, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37932457

RESUMEN

Regulatory T cells (Treg cells) are instrumental in establishing immunological tolerance. However, the precise effector mechanisms by which Treg cells control a specific type of immune response in a given tissue remains unresolved. By simultaneously studying Treg cells from different tissue origins under systemic autoimmunity, in the present study we show that interleukin (IL)-27 is specifically produced by intestinal Treg cells to regulate helper T17 cell (TH17 cell) immunity. Selectively increased intestinal TH17 cell responses in mice with Treg cell-specific IL-27 ablation led to exacerbated intestinal inflammation and colitis-associated cancer, but also helped protect against enteric bacterial infection. Furthermore, single-cell transcriptomic analysis has identified a CD83+CD62Llo Treg cell subset that is distinct from previously characterized intestinal Treg cell populations as the main IL-27 producers. Collectively, our study uncovers a new Treg cell suppression mechanism crucial for controlling a specific type of immune response in a particular tissue and provides further mechanistic insights into tissue-specific Treg cell-mediated immune regulation.


Asunto(s)
Interleucina-27 , Linfocitos T Reguladores , Ratones , Animales , Linfocitos T Colaboradores-Inductores , Tolerancia Inmunológica , Inmunidad Celular , Células Th17
2.
Immunity ; 57(9): 1995-1999, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39260349

RESUMEN

The immune system has a vital, albeit complex, relationship with the microbes residing within us, one that we are only beginning to understand. We asked investigators what they felt were the fundamental challenges we currently face in unraveling the impacts of microbes and their metabolites on host immunity and to discuss key opportunities toward achieving future insights and innovation.


Asunto(s)
Inmunidad , Humanos , Animales , Microbiota/inmunología , Interacciones Huésped-Patógeno/inmunología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Bacterias/inmunología , Bacterias/metabolismo
3.
Cell ; 163(2): 271-2, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26451475

RESUMEN

An effective immune response leads to rapid elimination of infectious agents, with seemingly little long-term impairment to the host. New findings by Morais da Fonseca et al. reveal that acute infections may result in permanent disruption of tissue homeostasis and immune dysfunction, long after clearance of a pathogen.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades del Sistema Inmune/microbiología , Enfermedades del Sistema Inmune/patología , Enfermedades Linfáticas/patología , Infecciones por Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/fisiología , Femenino , Humanos , Masculino
4.
Nature ; 626(7998): 419-426, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38052229

RESUMEN

Determining the structure and phenotypic context of molecules detected in untargeted metabolomics experiments remains challenging. Here we present reverse metabolomics as a discovery strategy, whereby tandem mass spectrometry spectra acquired from newly synthesized compounds are searched for in public metabolomics datasets to uncover phenotypic associations. To demonstrate the concept, we broadly synthesized and explored multiple classes of metabolites in humans, including N-acyl amides, fatty acid esters of hydroxy fatty acids, bile acid esters and conjugated bile acids. Using repository-scale analysis1,2, we discovered that some conjugated bile acids are associated with inflammatory bowel disease (IBD). Validation using four distinct human IBD cohorts showed that cholic acids conjugated to Glu, Ile/Leu, Phe, Thr, Trp or Tyr are increased in Crohn's disease. Several of these compounds and related structures affected pathways associated with IBD, such as interferon-γ production in CD4+ T cells3 and agonism of the pregnane X receptor4. Culture of bacteria belonging to the Bifidobacterium, Clostridium and Enterococcus genera produced these bile amidates. Because searching repositories with tandem mass spectrometry spectra has only recently become possible, this reverse metabolomics approach can now be used as a general strategy to discover other molecules from human and animal ecosystems.


Asunto(s)
Amidas , Ácidos y Sales Biliares , Ésteres , Ácidos Grasos , Metabolómica , Animales , Humanos , Bifidobacterium/metabolismo , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Clostridium/metabolismo , Estudios de Cohortes , Enfermedad de Crohn/metabolismo , Enterococcus/metabolismo , Ésteres/química , Ésteres/metabolismo , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Metabolómica/métodos , Fenotipo , Receptor X de Pregnano/metabolismo , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem , Amidas/química , Amidas/metabolismo
5.
Nat Immunol ; 14(7): 668-75, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23778794

RESUMEN

Pattern-recognition receptors (PRRs) are traditionally known to sense microbial molecules during infection to initiate inflammatory responses. However, ligands for PRRs are not exclusive to pathogens and are abundantly produced by the resident microbiota during normal colonization. Mechanism(s) that underlie this paradox have remained unclear. Recent studies reveal that gut bacterial ligands from the microbiota signal through PRRs to promote development of host tissue and the immune system, and protection from disease. Evidence from both invertebrate and vertebrate models reveals that innate immune receptors are required to promote long-term colonization by the microbiota. This emerging perspective challenges current models in immunology and suggests that PRRs may have evolved, in part, to mediate the bidirectional cross-talk between microbial symbionts and their hosts.


Asunto(s)
Tracto Gastrointestinal/microbiología , Inmunidad Innata/inmunología , Metagenoma/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Simbiosis/inmunología , Animales , Tracto Gastrointestinal/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos
6.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33472859

RESUMEN

The COVID-19 pandemic has the potential to affect the human microbiome in infected and uninfected individuals, having a substantial impact on human health over the long term. This pandemic intersects with a decades-long decline in microbial diversity and ancestral microbes due to hygiene, antibiotics, and urban living (the hygiene hypothesis). High-risk groups succumbing to COVID-19 include those with preexisting conditions, such as diabetes and obesity, which are also associated with microbiome abnormalities. Current pandemic control measures and practices will have broad, uneven, and potentially long-term effects for the human microbiome across the planet, given the implementation of physical separation, extensive hygiene, travel barriers, and other measures that influence overall microbial loss and inability for reinoculation. Although much remains uncertain or unknown about the virus and its consequences, implementing pandemic control practices could significantly affect the microbiome. In this Perspective, we explore many facets of COVID-19-induced societal changes and their possible effects on the microbiome, and discuss current and future challenges regarding the interplay between this pandemic and the microbiome. Recent recognition of the microbiome's influence on human health makes it critical to consider both how the microbiome, shaped by biosocial processes, affects susceptibility to the coronavirus and, conversely, how COVID-19 disease and prevention measures may affect the microbiome. This knowledge may prove key in prevention and treatment, and long-term biological and social outcomes of this pandemic.


Asunto(s)
COVID-19/microbiología , Hipótesis de la Higiene , Microbiota , Anciano , Antiinfecciosos/uso terapéutico , COVID-19/mortalidad , Ingestión de Alimentos , Femenino , Humanos , Lactante , Control de Infecciones/métodos , Masculino , Microbiota/efectos de los fármacos , Distanciamiento Físico , Embarazo
7.
BMC Cancer ; 23(1): 141, 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36765318

RESUMEN

BACKGROUND: The epidemiology and treatment of acute promyelocytic leukaemia (APL) are changing. We have incorporated oral arsenic trioxide (oral-ATO) into induction/maintenance. METHODS: Newly-diagnosed APL from 1991 to 2021 divided into three 10-year periods were studied to define its epidemiology and how oral-ATO impacted on its outcome. Primary endpoints included APL incidence, early deaths (ED, first 30 days), and overall survival (OS). Secondary endpoints included post-30-day OS, relapse-free survival (RFS), and incidence of second cancers. RESULTS: APL occurred in 374 males and 387 females at a median age of 44 (1-97) years. Annual incidences increased progressively, averaging 0.32 per 100,000 people. All-trans retinoic acid (ATRA)-based and oral-ATO-based regimens were used in 469 and 282 patients. There were 144 EDs, occurring almost exclusively in ATRA-based inductions (N = 139), being more with males, age > 50 years, leucocyte > 10 × 109/L, diagnosis during 1991-2009 and fewer with oral-ATO-based regimens. After a median of 75 (interquartile range: 14-161) months, 5-year and 10-year OS were 68.1% and 63.3%, inferior with males, age > 50 years, leucocyte > 10 × 109/L, high-risk Sanz score and superior with oral-ATO-based regimens. Factoring out EDs, 5-year and 10-year post-30-day OS were 84.0% and 78.1%, inferior with males and superior with oral-ATO-based regimens. In 607 CR1 patients, the 5-year RFS was 83.8%, superior with diagnosis in 2010-2021 and oral-ATO-based regimens. Second cancers developed in 21 patients, unrelated to oral-ATO-based regimens. CONCLUSIONS: There was an increasing incidence of APL, and all survivals were superior with the use of oral-ATO-based regimens. This study formed part of the Acute Promyelocytic Leukaemia Asian Consortium Project (ClinicalTrials.gov identifier: NCT04251754).


Asunto(s)
Arsenicales , Leucemia Promielocítica Aguda , Neoplasias Primarias Secundarias , Masculino , Femenino , Humanos , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Trióxido de Arsénico/efectos adversos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/epidemiología , Leucemia Promielocítica Aguda/diagnóstico , Recurrencia Local de Neoplasia , Tretinoina/efectos adversos , Resultado del Tratamiento , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Óxidos
8.
Nat Immunol ; 11(1): 76-83, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19855381

RESUMEN

Antimicrobial peptides are important effectors of innate immunity throughout the plant and animal kingdoms. In the mammalian small intestine, Paneth cell alpha-defensins are antimicrobial peptides that contribute to host defense against enteric pathogens. To determine if alpha-defensins also govern intestinal microbial ecology, we analyzed the intestinal microbiota of mice expressing a human alpha-defensin gene (DEFA5) and in mice lacking an enzyme required for the processing of mouse alpha-defensins. In these complementary models, we detected significant alpha-defensin-dependent changes in microbiota composition, but not in total bacterial numbers. Furthermore, DEFA5-expressing mice had striking losses of segmented filamentous bacteria and fewer interleukin 17 (IL-17)-producing lamina propria T cells. Our data ascribe a new homeostatic role to alpha-defensins in regulating the makeup of the commensal microbiota.


Asunto(s)
Ecología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , alfa-Defensinas/metabolismo , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/crecimiento & desarrollo , Recuento de Colonia Microbiana , Femenino , Citometría de Flujo , Humanos , Hibridación Fluorescente in Situ , Interleucina-17/inmunología , Interleucina-17/metabolismo , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Intestinos/inmunología , Masculino , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/metabolismo , Metagenoma , Ratones , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Filogenia , ARN Ribosómico 16S/genética , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , alfa-Defensinas/genética , alfa-Defensinas/inmunología
9.
Int J Mol Sci ; 23(10)2022 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-35628480

RESUMEN

In myelodysplastic syndrome (MDS), resistance to hypomethylating agents (HMA) portends a poor prognosis, underscoring the importance of understanding the molecular mechanisms leading to HMA-resistance. In this study, P39 and Kasumi-1 cells and their azacitidine-resistant and decitabine-resistant sublines were evaluated comparatively with transcriptomic and methylomic analyses. Expression profiling and genome-wide methylation microarray showed downregulation of PTEN associated with DNA hypermethylation in P39 cell lines resistant to azacitidine and decitabine. This pattern of PTEN dysregulation was also confirmed in a cohort of patients failing treatment with HMA. DNA hypomethylation of MDM2 was detected with downregulation of MDM2 in HMA resistant cell lines. Long-read sequencing revealed significant RNA hypomethylation of MDM2 resulting in alternative splicing and production of a truncated MDM2 transcript in azacitidine-resistant P39 cells. The expression of this MDM2 truncated transcript was also significantly increased in HMA-resistant patients compared with HMA-responsive patients. In conclusion, epigenetic and epi-transcriptomic dysregulation of PTEN and MDM2 were associated with resistance to hypomethylating agents.


Asunto(s)
Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Neoplasias Primarias Secundarias , Fosfohidrolasa PTEN , Proteínas Proto-Oncogénicas c-mdm2 , Azacitidina/farmacología , Azacitidina/uso terapéutico , Línea Celular Tumoral , Metilación de ADN , Decitabina/farmacología , Epigénesis Genética , Silenciador del Gen , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Neoplasias Primarias Secundarias/genética , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-mdm2/genética
10.
Int J Mol Sci ; 22(2)2021 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-33440869

RESUMEN

Myeloproliferative neoplasms (MPNs) are unique hematopoietic stem cell disorders sharing mutations that constitutively activate the signal-transduction pathways involved in haematopoiesis. They are characterized by stem cell-derived clonal myeloproliferation. The key MPNs comprise chronic myeloid leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). CML is defined by the presence of the Philadelphia (Ph) chromosome and BCR-ABL1 fusion gene. Despite effective cytoreductive agents and targeted therapy, complete CML/MPN stem cell eradication is rarely achieved. In this review article, we discuss the novel agents and combination therapy that can potentially abnormal hematopoietic stem cells in CML and MPNs and the CML/MPN stem cell-sustaining bone marrow microenvironment.


Asunto(s)
Susceptibilidad a Enfermedades , Células Madre Hematopoyéticas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/etiología , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Trastornos Mieloproliferativos/etiología , Trastornos Mieloproliferativos/terapia , Células Madre Neoplásicas/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Autofagia , Biomarcadores de Tumor , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Terapia Combinada , Predisposición Genética a la Enfermedad , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Terapia Molecular Dirigida , Trastornos Mieloproliferativos/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Cromosoma Filadelfia , Transducción de Señal/efectos de los fármacos , Nicho de Células Madre , Microambiente Tumoral
11.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638574

RESUMEN

Myelodysplastic syndrome (MDS) is a heterogeneous, clonal hematological disorder characterized by ineffective hematopoiesis, cytopenia, morphologic dysplasia, and predisposition to acute myeloid leukemia (AML). Stem cell genomic instability, microenvironmental aberrations, and somatic mutations contribute to leukemic transformation. The hypomethylating agents (HMAs), azacitidine and decitabine are the standard of care for patients with higher-risk MDS. Although these agents induce responses in up to 40-60% of patients, primary or secondary drug resistance is relatively common. To improve the treatment outcome, combinational therapies comprising HMA with targeted therapy or immunotherapy are being evaluated and are under continuous development. This review provides a comprehensive update of the molecular pathogenesis and immune-dysregulations involved in MDS, mechanisms of resistance to HMA, and strategies to overcome HMA resistance.


Asunto(s)
Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/terapia , Antineoplásicos/uso terapéutico , Terapia Combinada/métodos , Metilación de ADN/efectos de los fármacos , Humanos , Inmunoterapia/métodos , Terapia Molecular Dirigida/métodos , Síndromes Mielodisplásicos/inmunología , Resultado del Tratamiento
12.
Gastroenterology ; 156(8): 2174-2189, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30880022

RESUMEN

The intestinal microbiota is a dynamic community of bacteria, fungi, and viruses that mediates mucosal homeostasis and physiology. Imbalances in the microbiome and aberrant immune responses to gut bacteria can disrupt homeostasis and are associated with inflammatory bowel diseases (IBDs) in humans and colitis in mice. We review genetic variants associated with IBD and their effects on the intestinal microbiome, the immune response, and disease pathogenesis. The intestinal microbiome, which includes microbial antigens, adjuvants, and metabolic products, affects the development and function of the intestinal mucosa, influencing inflammatory responses in the gut. Therefore, strategies to manipulate the microbiome might be used in treatment of IBD. We review microbe-based therapies for IBD and the potential to engineer patients' intestinal microbiota. We discuss how studies of patients with IBD and mouse models have advanced our understanding of the interactions between genetic factors and the gut microbiome, and challenges to the development of microbe-based therapies for IBD.


Asunto(s)
Drogas en Investigación/administración & dosificación , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Inmunoterapia/métodos , Enfermedades Inflamatorias del Intestino/terapia , Guías de Práctica Clínica como Asunto , Animales , Drogas en Investigación/farmacología , Genoma Humano , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Ratones , Modelos Animales , Pronóstico , Resultado del Tratamiento
14.
Nat Rev Immunol ; 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39009868

RESUMEN

Bile acids are increasingly appearing in the spotlight owing to their novel impacts on various host processes. Similarly, there is growing attention on members of the microbiota that are responsible for bile acid modifications. With recent advances in technology enabling the discovery and continued identification of microbially conjugated bile acids, the chemical complexity of the bile acid landscape in the body is increasing at a rapid pace. In this Review, we summarize our current understanding of how bile acids and the gut microbiota interact to modulate immune responses during homeostasis and disease, with a particular focus on the gut.

15.
Cell Host Microbe ; 32(2): 181-190.e9, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38228143

RESUMEN

The early microbial colonization of the gastrointestinal tract can have long-term impacts on development and health. Keystone species, including Bacteroides spp., are prominent in early life and play crucial roles in maintaining the structure of the intestinal ecosystem. However, the process by which a resilient community is curated during early life remains inadequately understood. Here, we show that a single sialidase, NanH, in Bacteroides fragilis mediates stable occupancy of the intestinal mucosa in early life and regulates a commensal colonization program. This program is triggered by sialylated glycans, including those found in human milk oligosaccharides and intestinal mucus. NanH is required for vertical transmission from dams to pups and promotes B. fragilis dominance during early life. Furthermore, NanH facilitates commensal resilience and recovery after antibiotic treatment in a defined microbial community. Collectively, our study reveals a co-evolutionary mechanism between the host and microbiota mediated through host-derived glycans to promote stable colonization.


Asunto(s)
Ecosistema , Neuraminidasa , Humanos , Bacteroides fragilis , Mucosa Intestinal/microbiología , Polisacáridos
16.
mSystems ; 9(7): e0051624, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38934546

RESUMEN

Bacteroides fragilis is a Gram-negative commensal bacterium commonly found in the human colon, which differentiates into two genomospecies termed divisions I and II. Through a comprehensive collection of 694 B. fragilis whole genome sequences, we identify novel features distinguishing these divisions. Our study reveals a distinct geographic distribution with division I strains predominantly found in North America and division II strains in Asia. Additionally, division II strains are more frequently associated with bloodstream infections, suggesting a distinct pathogenic potential. We report differences between the two divisions in gene abundance related to metabolism, virulence, stress response, and colonization strategies. Notably, division II strains harbor more antimicrobial resistance (AMR) genes than division I strains. These findings offer new insights into the functional roles of division I and II strains, indicating specialized niches within the intestine and potential pathogenic roles in extraintestinal sites. IMPORTANCE: Understanding the distinct functions of microbial species in the gut microbiome is crucial for deciphering their impact on human health. Classifying division II strains as Bacteroides fragilis can lead to erroneous associations, as researchers may mistakenly attribute characteristics observed in division II strains to the more extensively studied division I B. fragilis. Our findings underscore the necessity of recognizing these divisions as separate species with distinct functions. We unveil new findings of differential gene prevalence between division I and II strains in genes associated with intestinal colonization and survival strategies, potentially influencing their role as gut commensals and their pathogenicity in extraintestinal sites. Despite the significant niche overlap and colonization patterns between these groups, our study highlights the complex dynamics that govern strain distribution and behavior, emphasizing the need for a nuanced understanding of these microorganisms.


Asunto(s)
Bacteroides fragilis , Variación Genética , Genoma Bacteriano , Bacteroides fragilis/genética , Bacteroides fragilis/patogenicidad , Bacteroides fragilis/aislamiento & purificación , Humanos , Genoma Bacteriano/genética , Microbioma Gastrointestinal/genética , Filogenia , Infecciones por Bacteroides/microbiología , Secuenciación Completa del Genoma , Farmacorresistencia Bacteriana/genética
17.
J Exp Med ; 221(1)2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38085267

RESUMEN

Type I interferons (IFNs) exert a broad range of biological effects important in coordinating immune responses, which have classically been studied in the context of pathogen clearance. Yet, whether immunomodulatory bacteria operate through IFN pathways to support intestinal immune tolerance remains elusive. Here, we reveal that the commensal bacterium, Bacteroides fragilis, utilizes canonical antiviral pathways to modulate intestinal dendritic cells (DCs) and regulatory T cell (Treg) responses. Specifically, IFN signaling is required for commensal-induced tolerance as IFNAR1-deficient DCs display blunted IL-10 and IL-27 production in response to B. fragilis. We further establish that IFN-driven IL-27 in DCs is critical in shaping the ensuing Foxp3+ Treg via IL-27Rα signaling. Consistent with these findings, single-cell RNA sequencing of gut Tregs demonstrated that colonization with B. fragilis promotes a distinct IFN gene signature in Foxp3+ Tregs during intestinal inflammation. Altogether, our findings demonstrate a critical role of commensal-mediated immune tolerance via tonic type I IFN signaling.


Asunto(s)
Interferón Tipo I , Interleucina-27 , Ratones , Animales , Interleucina-27/metabolismo , Linfocitos T Reguladores , Interferón Tipo I/metabolismo , Tolerancia Inmunológica , Factores de Transcripción Forkhead/metabolismo , Bacterias/metabolismo , Células Dendríticas
18.
bioRxiv ; 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38948766

RESUMEN

Bacteroides fragilis is a prominent member of the human gut microbiota, playing crucial roles in maintaining gut homeostasis and host health. Although it primarily functions as a beneficial commensal, B. fragilis can become pathogenic. To determine the genetic basis of its duality, we conducted a comparative genomic analysis of 813 B. fragilis strains, representing both commensal and pathogenic origins. Our findings reveal that pathogenic strains emerge across diverse phylogenetic lineages, due in part to rapid gene exchange and the adaptability of the accessory genome. We identified 16 phylogenetic groups, differentiated by genes associated with capsule composition, interspecies competition, and host interactions. A microbial genome-wide association study identified 44 genes linked to extra-intestinal survival and pathogenicity. These findings reveal how genomic diversity within commensal species can lead to the emergence of pathogenic traits, broadening our understanding of microbial evolution in the gut.

19.
Nat Microbiol ; 9(2): 336-345, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38316926

RESUMEN

microbeMASST, a taxonomically informed mass spectrometry (MS) search tool, tackles limited microbial metabolite annotation in untargeted metabolomics experiments. Leveraging a curated database of >60,000 microbial monocultures, users can search known and unknown MS/MS spectra and link them to their respective microbial producers via MS/MS fragmentation patterns. Identification of microbe-derived metabolites and relative producers without a priori knowledge will vastly enhance the understanding of microorganisms' role in ecology and human health.


Asunto(s)
Metabolómica , Espectrometría de Masas en Tándem , Humanos , Metabolómica/métodos , Bases de Datos Factuales
20.
bioRxiv ; 2023 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-37609270

RESUMEN

The early microbial colonization of the gastrointestinal tract can lead to long-term impacts in development and overall human health. Keystone species, including Bacteroides spp ., play a crucial role in maintaining the structure, diversity, and function of the intestinal ecosystem. However, the process by which a defined and resilient community is curated and maintained during early life remains inadequately understood. Here, we show that a single sialidase, NanH, in Bacteroides fragilis mediates stable occupancy of the intestinal mucosa and regulates the commensal colonization program during the first weeks of life. This program is triggered by sialylated glycans, including those found in human milk oligosaccharides and intestinal mucus. After examining the dynamics between pioneer gut Bacteroides species in the murine gut, we discovered that NanH enables vertical transmission from dams to pups and promotes B. fragilis dominance during early life. Furthermore, we demonstrate that NanH facilitates commensal resilience and recovery after antibiotic treatment in a defined microbial community. Collectively, our study reveals a co-evolutionary mechanism between the host and the microbiota mediated through host-derived glycans to promote stable intestinal colonization.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA