Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(3): 547-562.e22, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35051369

RESUMEN

Hundreds of microbiota genes are associated with host biology/disease. Unraveling the causal contribution of a microbiota gene to host biology remains difficult because many are encoded by nonmodel gut commensals and not genetically targetable. A general approach to identify their gene transfer methodology and build their gene manipulation tools would enable mechanistic dissections of their impact on host physiology. We developed a pipeline that identifies the gene transfer methods for multiple nonmodel microbes spanning five phyla, and we demonstrated the utility of their genetic tools by modulating microbiome-derived short-chain fatty acids and bile acids in vitro and in the host. In a proof-of-principle study, by deleting a commensal gene for bile acid synthesis in a complex microbiome, we discovered an intriguing role of this gene in regulating colon inflammation. This technology will enable genetically engineering the nonmodel gut microbiome and facilitate mechanistic dissection of microbiota-host interactions.


Asunto(s)
Microbioma Gastrointestinal/genética , Genes Bacterianos , Animales , Ácidos y Sales Biliares/metabolismo , Sistemas CRISPR-Cas/genética , Clostridium/genética , Colitis/inducido químicamente , Colitis/microbiología , Colitis/patología , Sulfato de Dextran , Farmacorresistencia Microbiana/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Técnicas de Transferencia de Gen , Vida Libre de Gérmenes , Inflamación/patología , Intestinos/patología , Masculino , Metaboloma/genética , Metagenómica , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Insercional/genética , Mutación/genética , ARN Ribosómico 16S/genética , Transcripción Genética
2.
Cell ; 185(22): 4170-4189.e20, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36240781

RESUMEN

Nociceptive pain is a hallmark of many chronic inflammatory conditions including inflammatory bowel diseases (IBDs); however, whether pain-sensing neurons influence intestinal inflammation remains poorly defined. Employing chemogenetic silencing, adenoviral-mediated colon-specific silencing, and pharmacological ablation of TRPV1+ nociceptors, we observed more severe inflammation and defective tissue-protective reparative processes in a murine model of intestinal damage and inflammation. Disrupted nociception led to significant alterations in the intestinal microbiota and a transmissible dysbiosis, while mono-colonization of germ-free mice with Gram+Clostridium spp. promoted intestinal tissue protection through a nociceptor-dependent pathway. Mechanistically, disruption of nociception resulted in decreased levels of substance P, and therapeutic delivery of substance P promoted tissue-protective effects exerted by TRPV1+ nociceptors in a microbiota-dependent manner. Finally, dysregulated nociceptor gene expression was observed in intestinal biopsies from IBD patients. Collectively, these findings indicate an evolutionarily conserved functional link between nociception, the intestinal microbiota, and the restoration of intestinal homeostasis.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Ratones , Animales , Microbioma Gastrointestinal/fisiología , Nociceptores/fisiología , Sustancia P , Disbiosis , Inflamación
3.
Nat Immunol ; 24(11): 1879-1889, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37872315

RESUMEN

Gastrointestinal fungal dysbiosis is a hallmark of several diseases marked by systemic immune activation. Whether persistent pathobiont colonization during immune alterations and impaired gut barrier function has a durable impact on host immunity is unknown. We found that elevated levels of Candida albicans immunoglobulin G (IgG) antibodies marked patients with severe COVID-19 (sCOVID-19) who had intestinal Candida overgrowth, mycobiota dysbiosis and systemic neutrophilia. Analysis of hematopoietic stem cell progenitors in sCOVID-19 revealed transcriptional changes in antifungal immunity pathways and reprogramming of granulocyte myeloid progenitors (GMPs) for up to a year. Mice colonized with C. albicans patient isolates experienced increased lung neutrophilia and pulmonary NETosis during severe acute respiratory syndrome coronavirus-2 infection, which were partially resolved with antifungal treatment or by interleukin-6 receptor blockade. sCOVID-19 patients treated with tocilizumab experienced sustained reductions in C. albicans IgG antibodies titers and GMP transcriptional changes. These findings suggest that gut fungal pathobionts may contribute to immune activation during inflammatory diseases, offering potential mycobiota-immune therapeutic strategies for sCOVID-19 with prolonged symptoms.


Asunto(s)
COVID-19 , Micobioma , Humanos , Animales , Ratones , Antifúngicos , Disbiosis , Neutrófilos , Candida albicans , Inmunoglobulina G
4.
Immunity ; 57(1): 14-27, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38198849

RESUMEN

Nutrition profoundly shapes immunity and inflammation across the lifespan of mammals, from pre- and post-natal periods to later life. Emerging insights into diet-microbiota interactions indicate that nutrition has a dominant influence on the composition-and metabolic output-of the intestinal microbiota, which in turn has major consequences for host immunity and inflammation. Here, we discuss recent findings that support the concept that dietary effects on microbiota-derived metabolites potently alter immune responses in health and disease. We discuss how specific dietary components and metabolites can be either pro-inflammatory or anti-inflammatory in a context- and tissue-dependent manner during infection, chronic inflammation, and cancer. Together, these studies emphasize the influence of diet-microbiota crosstalk on immune regulation that will have a significant impact on precision nutrition approaches and therapeutic interventions for managing inflammation, infection, and cancer immunotherapy.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Neoplasias , Animales , Inflamación , Reacciones Cruzadas , Neoplasias/terapia , Mamíferos
5.
Immunity ; 57(4): 876-889.e11, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38479384

RESUMEN

Concentrations of the secondary bile acid, deoxycholic acid (DCA), are aberrantly elevated in colorectal cancer (CRC) patients, but the consequences remain poorly understood. Here, we screened a library of gut microbiota-derived metabolites and identified DCA as a negative regulator for CD8+ T cell effector function. Mechanistically, DCA suppressed CD8+ T cell responses by targeting plasma membrane Ca2+ ATPase (PMCA) to inhibit Ca2+-nuclear factor of activated T cells (NFAT)2 signaling. In CRC patients, CD8+ T cell effector function negatively correlated with both DCA concentration and expression of a bacterial DCA biosynthetic gene. Bacteria harboring DCA biosynthetic genes suppressed CD8+ T cells effector function and promoted tumor growth in mice. This effect was abolished by disrupting bile acid metabolism via bile acid chelation, genetic ablation of bacterial DCA biosynthetic pathway, or specific bacteriophage. Our study demonstrated causation between microbial DCA metabolism and anti-tumor CD8+ T cell response in CRC, suggesting potential directions for anti-tumor therapy.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Humanos , Ratones , Animales , Ácidos y Sales Biliares , Ácido Desoxicólico/farmacología , Linfocitos T CD8-positivos
6.
Cell ; 168(3): 517-526.e18, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28111075

RESUMEN

The gut microbiota modulate host biology in numerous ways, but little is known about the molecular mediators of these interactions. Previously, we found a widely distributed family of nonribosomal peptide synthetase gene clusters in gut bacteria. Here, by expressing a subset of these clusters in Escherichia coli or Bacillus subtilis, we show that they encode pyrazinones and dihydropyrazinones. At least one of the 47 clusters is present in 88% of the National Institutes of Health Human Microbiome Project (NIH HMP) stool samples, and they are transcribed under conditions of host colonization. We present evidence that the active form of these molecules is the initially released peptide aldehyde, which bears potent protease inhibitory activity and selectively targets a subset of cathepsins in human cell proteomes. Our findings show that an approach combining bioinformatics, synthetic biology, and heterologous gene cluster expression can rapidly expand our knowledge of the metabolic potential of the microbiota while avoiding the challenges of cultivating fastidious commensals.


Asunto(s)
Bacterias/metabolismo , Microbioma Gastrointestinal , Microbiota , Péptido Sintasas/metabolismo , Pirazinas/metabolismo , Animales , Bacillus subtilis/genética , Bacterias/clasificación , Bacterias/genética , Escherichia coli/genética , Heces/microbiología , Humanos , Péptido Sintasas/genética , Filogenia
7.
Nature ; 611(7936): 578-584, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36323778

RESUMEN

Dietary fibres can exert beneficial anti-inflammatory effects through microbially fermented short-chain fatty acid metabolites<sup>1,2</sup>, although the immunoregulatory roles of most fibre diets and their microbiota-derived metabolites remain poorly defined. Here, using microbial sequencing and untargeted metabolomics, we show that a diet of inulin fibre alters the composition of the mouse microbiota and the levels of microbiota-derived metabolites, notably bile acids. This metabolomic shift is associated with type 2 inflammation in the intestine and lungs, characterized by IL-33 production, activation of group 2 innate lymphoid cells and eosinophilia. Delivery of cholic acid mimics inulin-induced type 2 inflammation, whereas deletion of the bile acid receptor farnesoid X receptor diminishes the effects of inulin. The effects of inulin are microbiota dependent and were reproduced in mice colonized with human-derived microbiota. Furthermore, genetic deletion of a bile-acid-metabolizing enzyme in one bacterial species abolishes the ability of inulin to trigger type 2 inflammation. Finally, we demonstrate that inulin enhances allergen- and helminth-induced type 2 inflammation. Taken together, these data reveal that dietary inulin fibre triggers microbiota-derived cholic acid and type 2 inflammation at barrier surfaces with implications for understanding the pathophysiology of allergic inflammation, tissue protection and host defence.


Asunto(s)
Ácidos y Sales Biliares , Fibras de la Dieta , Microbioma Gastrointestinal , Inflamación , Inulina , Animales , Humanos , Ratones , Ácidos y Sales Biliares/metabolismo , Ácido Cólico/farmacología , Fibras de la Dieta/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Inmunidad Innata , Inflamación/inducido químicamente , Inflamación/clasificación , Inflamación/patología , Inulina/farmacología , Linfocitos/citología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Metabolómica , Pulmón/efectos de los fármacos , Pulmón/patología , Intestinos/efectos de los fármacos , Intestinos/microbiología , Intestinos/patología , Interleucina-33/metabolismo , Eosinófilos/citología , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología
8.
Nature ; 582(7813): 566-570, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32555455

RESUMEN

The gut microbiota synthesize hundreds of molecules, many of which influence host physiology. Among the most abundant metabolites are the secondary bile acids deoxycholic acid (DCA) and lithocholic acid (LCA), which accumulate at concentrations of around 500 µM and are known to block the growth of Clostridium difficile1, promote hepatocellular carcinoma2 and modulate host metabolism via the G-protein-coupled receptor TGR5 (ref. 3). More broadly, DCA, LCA and their derivatives are major components of the recirculating pool of bile acids4; the size and composition of this pool are a target of therapies for primary biliary cholangitis and nonalcoholic steatohepatitis. Nonetheless, despite the clear impact of DCA and LCA on host physiology, an incomplete knowledge of their biosynthetic genes and a lack of genetic tools to enable modification of their native microbial producers limit our ability to modulate secondary bile acid levels in the host. Here we complete the pathway to DCA and LCA by assigning and characterizing enzymes for each of the steps in its reductive arm, revealing a strategy in which the A-B rings of the steroid core are transiently converted into an electron acceptor for two reductive steps carried out by Fe-S flavoenzymes. Using anaerobic in vitro reconstitution, we establish that a set of six enzymes is necessary and sufficient for the eight-step conversion of cholic acid to DCA. We then engineer the pathway into Clostridium sporogenes, conferring production of DCA and LCA on a nonproducing commensal and demonstrating that a microbiome-derived pathway can be expressed and controlled heterologously. These data establish a complete pathway to two central components of the bile acid pool.


Asunto(s)
Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Hidroxilación/genética , Redes y Vías Metabólicas/genética , Animales , Clostridium/enzimología , Clostridium/genética , Clostridium/metabolismo , Ácido Desoxicólico/química , Ácido Desoxicólico/metabolismo , Ácido Litocólico/química , Ácido Litocólico/metabolismo , Masculino , Ingeniería Metabólica , Ratones , Operón/genética , Simbiosis
9.
Nature ; 581(7809): 475-479, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32461639

RESUMEN

Intestinal health relies on the immunosuppressive activity of CD4+ regulatory T (Treg) cells1. Expression of the transcription factor Foxp3 defines this lineage, and can be induced extrathymically by dietary or commensal-derived antigens in a process assisted by a Foxp3 enhancer known as conserved non-coding sequence 1 (CNS1)2-4. Products of microbial fermentation including butyrate facilitate the generation of peripherally induced Treg (pTreg) cells5-7, indicating that metabolites shape the composition of the colonic immune cell population. In addition to dietary components, bacteria modify host-derived molecules, generating a number of biologically active substances. This is epitomized by the bacterial transformation of bile acids, which creates a complex pool of steroids8 with a range of physiological functions9. Here we screened the major species of deconjugated bile acids for their ability to potentiate the differentiation of pTreg cells. We found that the secondary bile acid 3ß-hydroxydeoxycholic acid (isoDCA) increased Foxp3 induction by acting on dendritic cells (DCs) to diminish their immunostimulatory properties. Ablating one receptor, the farnesoid X receptor, in DCs enhanced the generation of Treg cells and imposed a transcriptional profile similar to that induced by isoDCA, suggesting an interaction between this bile acid and nuclear receptor. To investigate isoDCA in vivo, we took a synthetic biology approach and designed minimal microbial consortia containing engineered Bacteroides strains. IsoDCA-producing consortia increased the number of colonic RORγt-expressing Treg cells in a CNS1-dependent manner, suggesting enhanced extrathymic differentiation.


Asunto(s)
Bacterias/metabolismo , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Secuencia de Aminoácidos , Animales , Bacteroides/metabolismo , Colon/microbiología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Fermentación , Microbioma Gastrointestinal , Masculino , Ratones , Ratones Endogámicos C57BL , Consorcios Microbianos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo
10.
Gastroenterology ; 162(1): 166-178, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34606847

RESUMEN

BACKGROUND & AIMS: Fecal microbiota transplantation (FMT) is an emerging treatment modality for ulcerative colitis (UC). Several randomized controlled trials have shown efficacy for FMT in the treatment of UC, but a better understanding of the transferable microbiota and their immune impact is needed to develop more efficient microbiome-based therapies for UC. METHODS: Metagenomic analysis and strain tracking was performed on 60 donor and recipient samples receiving FMT for active UC. Sorting and sequencing of immunoglobulin (Ig) A-coated microbiota (called IgA-seq) was used to define immune-reactive microbiota. Colonization of germ-free or genetically engineered mice with patient-derived strains was performed to determine the mechanism of microbial impact on intestinal immunity. RESULTS: Metagenomic analysis defined a core set of donor-derived transferable bacterial strains in UC subjects achieving clinical response, which predicted response in an independent trial of FMT for UC. IgA-seq of FMT recipient samples and gnotobiotic mice colonized with donor microbiota identified Odoribacter splanchnicus as a transferable strain shaping mucosal immunity, which correlated with clinical response and the induction of mucosal regulatory T cells. Colonization of mice with O splanchnicus led to an increase in Foxp3+/RORγt+ regulatory T cells, induction of interleukin (IL) 10, and production of short chain fatty acids, all of which were required for O splanchnicus to limit colitis in mouse models. CONCLUSIONS: This work provides the first evidence of transferable, donor-derived strains that correlate with clinical response to FMT in UC and reveals O splanchnicus as a key component promoting both metabolic and immune cell protection from colitis. These mechanistic features will help enable strategies to enhance the efficacy of microbial therapy for UC. Clinicaltrials.gov ID NCT02516384.


Asunto(s)
Bacteroidetes/inmunología , Colitis/terapia , Colon/microbiología , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Inmunoglobulina A/inmunología , Mucosa Intestinal/microbiología , Animales , Bacteroidetes/genética , Bacteroidetes/metabolismo , Ensayos Clínicos como Asunto , Colitis/inmunología , Colitis/metabolismo , Colitis/microbiología , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/microbiología , Colon/inmunología , Colon/metabolismo , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Vida Libre de Gérmenes , Humanos , Inmunidad Mucosa , Inmunoglobulina A/genética , Inmunoglobulina A/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Linfocitos Intraepiteliales/microbiología , Metagenoma , Metagenómica , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/microbiología , Resultado del Tratamiento
11.
Circ Res ; 123(10): 1164-1176, 2018 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-30359185

RESUMEN

RATIONALE: Gut microbes influence cardiovascular disease and thrombosis risks through the production of trimethylamine N-oxide (TMAO). Microbiota-dependent generation of trimethylamine (TMA)-the precursor to TMAO-is rate limiting in the metaorganismal TMAO pathway in most humans and is catalyzed by several distinct microbial choline TMA-lyases, including the proteins encoded by the cutC/D (choline utilization C/D) genes in multiple human commensals. OBJECTIVE: Direct demonstration that the gut microbial cutC gene is sufficient to transmit enhanced platelet reactivity and thrombosis potential in a host via TMA/TMAO generation has not yet been reported. METHODS AND RESULTS: Herein, we use gnotobiotic mice and a series of microbial colonization studies to show that microbial cutC-dependent TMA/TMAO production is sufficient to transmit heightened platelet reactivity and thrombosis potential in a host. Specifically, we examine in vivo thrombosis potential employing germ-free mice colonized with either high TMA-producing stable human fecal polymcrobial communities or a defined CutC-deficient background microbial community coupled with a CutC-expressing human commensal±genetic disruption of its cutC gene (ie, Clostridium sporogenes Δ cutC). CONCLUSIONS: Collectively, these studies point to the microbial choline TMA-lyase pathway as a rational molecular target for the treatment of atherothrombotic heart disease.


Asunto(s)
Proteínas Bacterianas/metabolismo , Trasplante de Microbiota Fecal , Liasas/metabolismo , Activación Plaquetaria , Trombosis/microbiología , Adulto , Animales , Proteínas Bacterianas/genética , Colina/metabolismo , Clostridium/enzimología , Clostridium/genética , Femenino , Microbioma Gastrointestinal , Humanos , Liasas/genética , Masculino , Metilaminas/metabolismo , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Trombosis/sangre
12.
Mol Microbiol ; 103(2): 347-365, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27775185

RESUMEN

Fungal secondary metabolites (SMs) are extremely important in medicine and agriculture, but regulation of their biosynthesis is incompletely understood. We have developed a genetic screen in Aspergillus nidulans for negative regulators of fungal SM gene clusters and we have used this screen to isolate mutations that upregulate transcription of the non-ribosomal peptide synthetase gene required for nidulanin A biosynthesis. Several of these mutations are allelic and we have identified the mutant gene by genome sequencing. The gene, which we designate mcrA, is conserved but uncharacterized, and it encodes a putative transcription factor. Metabolite profiles of mcrA deletant, mcrA overexpressing, and parental strains reveal that mcrA regulates at least ten SM gene clusters. Deletion of mcrA stimulates SM production even in strains carrying a deletion of the SM regulator laeA, and deletion of mcrA homologs in Aspergillus terreus and Penicillum canescens alters the secondary metabolite profile of these organisms. Deleting mcrA in a genetic dereplication strain has allowed us to discover two novel compounds as well as an antibiotic not known to be produced by A. nidulans. Deletion of mcrA upregulates transcription of hundreds of genes including many that are involved in secondary metabolism, while downregulating a smaller number of genes.


Asunto(s)
Aspergillus nidulans/genética , Aspergillus nidulans/metabolismo , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica , Genes Fúngicos , Familia de Multigenes , Mutación , Metabolismo Secundario , Eliminación de Secuencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
13.
Fungal Genet Biol ; 119: 1-6, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30096370

RESUMEN

Acetylaranotin is an epipolythiodiketopiperazine (ETP) secondary metabolite with a broad range of bioactivities. We demonstrated that ATEG_01465.1 located outside of acetylaranotin gene cluster is responsible for catalyzing the S-methylation of its biosynthetic pathway. Combining the previous characterization of acetylaranotin biosynthetic gene cluster together with the identification of its S-methyltransferase provides a means to obtain second-generation acetylaranotin derivatives previously inaccessible. By permutations of targeted deletions of ATEG_01465.1, acetyltransferase (AtaH), and benzoate hydroxylase (AtaY), three novel acetylaranotin derivatives were produced by Aspergillus terreus.


Asunto(s)
Vías Biosintéticas/genética , Metiltransferasas/genética , Oxepinas/metabolismo , Piperazinas/metabolismo , Acetiltransferasas/genética , Aspergillus/genética , Aspergillus/metabolismo , Genoma Fúngico/genética , Familia de Multigenes/genética , Oxigenasas/genética , Eliminación de Secuencia/genética
14.
Fungal Genet Biol ; 89: 84-88, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26851300

RESUMEN

Genome sequencing of the fungus Aspergillus terreus uncovered a number of silent core structural biosynthetic genes encoding enzymes presumed to be involved in the production of cryptic secondary metabolites. There are five nonribosomal peptide synthetase (NRPS)-like genes with the predicted A-T-TE domain architecture within the A. terreus genome. Among the five genes, only the product of pgnA remains unknown. The Tet-on system is an inducible, tunable and metabolism-independent expression system originally developed for Aspergillus niger. Here we report the adoption of the Tet-on system as an effective gene activation tool in A. terreus. Application of this system in A. terreus allowed us to uncover the product of the cryptic NRPS-like gene, pgnA. Furthermore expression of pgnA in the heterologous Aspergillus nidulans host suggested that the pgnA gene alone is necessary for phenguignardic acid (1) biosynthesis.


Asunto(s)
Aspergillus/enzimología , Aspergillus/genética , Doxiciclina/farmacología , Genes Fúngicos/genética , Péptido Sintasas/genética , Aspergillus/efectos de los fármacos , Aspergillus/metabolismo , Aspergillus nidulans/efectos de los fármacos , Aspergillus nidulans/genética , Aspergillus niger/efectos de los fármacos , Aspergillus niger/genética , Productos Biológicos/metabolismo , Dioxolanos/metabolismo , Familia de Multigenes , Activación Transcripcional
15.
Proc Natl Acad Sci U S A ; 110(42): 17065-70, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24082142

RESUMEN

The hallmark trait of fungal secondary-metabolite gene clusters is well established, consisting of contiguous enzymatic and often regulatory gene(s) devoted to the production of a metabolite of a specific chemical class. Unexpectedly, we have found a deviation from this motif in a subtelomeric region of Aspergillus fumigatus. This region, under the control of the master regulator of secondary metabolism, LaeA, contains, in its entirety, the genetic machinery for three natural products (fumitremorgin, fumagillin, and pseurotin), where genes for fumagillin and pseurotin are physically intertwined in a single supercluster. Deletions of 29 adjoining genes revealed that fumagillin and pseurotin are coregulated by the supercluster-embedded regulatory gene with biosynthetic genes belonging to one of the two metabolic pathways in a noncontiguous manner. Comparative genomics indicates the fumagillin/pseurotin supercluster is maintained in a rapidly evolving region of diverse fungal genomes. This blended design confounds predictions from established secondary-metabolite cluster search algorithms and provides an expanded view of natural product evolution.


Asunto(s)
Aspergillus fumigatus/metabolismo , Ciclohexanos/metabolismo , Ácidos Grasos Insaturados/metabolismo , Genes Fúngicos/fisiología , Indenos/metabolismo , Familia de Multigenes/fisiología , Pirrolidinonas/metabolismo , Algoritmos , Aspergillus fumigatus/genética , Ácidos Grasos Insaturados/genética , Análisis de Secuencia de ADN/métodos , Sesquiterpenos/metabolismo
16.
PLoS Pathog ; 9(4): e1003289, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23592999

RESUMEN

The secondary metabolome provides pathogenic fungi with a plethoric and versatile panel of molecules that can be deployed during host ingress. While powerful genetic and analytical chemistry methods have been developed to identify fungal secondary metabolites (SMs), discovering the biological activity of SMs remains an elusive yet critical task. Here, we describe a process for identifying the immunosuppressive properties of Aspergillus SMs developed by coupling a cost-effective microfluidic neutrophil chemotaxis assay with an in vivo zebrafish assay. The microfluidic platform allows the identification of metabolites inhibiting neutrophil recruitment with as little as several nano-grams of compound in microliters of fluid. The zebrafish assay demonstrates a simple and accessible approach for performing in vivo studies without requiring any manipulation of the fish. Using this methodology we identify the immunosuppressive properties of a fungal SM, endocrocin. We find that endocrocin is localized in Aspergillus fumigatus spores and its biosynthesis is temperature-dependent. Finally, using the Drosophila toll deficient model, we find that deletion of encA, encoding the polyketide synthase required for endocrocin production, yields a less pathogenic strain of A. fumigatus when spores are harvested from endocrocin permissive but not when harvested from endocrocin restrictive conditions. The tools developed here will open new "function-omic" avenues downstream of the metabolomics, identification, and purification phases.


Asunto(s)
Aspergillus fumigatus/metabolismo , Aspergillus fumigatus/patogenicidad , Neutrófilos/inmunología , Animales , Antracenos/metabolismo , Aspergillus fumigatus/genética , Drosophila/enzimología , Drosophila/genética , Drosophila/inmunología , Terapia de Inmunosupresión , Metaboloma , Microfluídica , Neutrófilos/efectos de los fármacos , Sintasas Poliquetidas/genética , Sintasas Poliquetidas/metabolismo , Metabolismo Secundario , Pez Cebra/inmunología , Pez Cebra/metabolismo
17.
Molecules ; 20(1): 325-34, 2014 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-25547729

RESUMEN

Six sesquiterpenoids 1-6, including two new ones, an ent-daucane-type sesquiterpenoid, asperaculane A (1), and a nordaucane one, asperaculane B (2), and four known nordaucane derivatives, aculenes A-D 3-6, together with the known secalonic acid D (7), were isolated from a fermentation culture of the fungus Aspergillus aculeatus. Their structures and absolute configurations were established by analyses of their spectroscopic data, including 1D and 2D-NMR spectra, HR-ESIMS, electronic circular dichroism (ECD) data, and quantum chemical calculations. These metabolites were evaluated for in vitro cytotoxic activity against two cell lines, human cancer cell lines (HeLa) and one normal hamster cell line (CHO).


Asunto(s)
Aspergillus/química , Sesquiterpenos/aislamiento & purificación , Animales , Células CHO , Línea Celular Tumoral , Dicroismo Circular , Cricetinae , Cricetulus , Humanos , Espectroscopía de Resonancia Magnética , Sesquiterpenos/química , Sesquiterpenos/farmacología , Espectrometría de Masa por Ionización de Electrospray
18.
Cell Host Microbe ; 32(5): 661-675.e10, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38657606

RESUMEN

The intestine and liver are thought to metabolize dietary nutrients and regulate host nutrient homeostasis. Here, we find that the gut microbiota also reshapes the host amino acid (aa) landscape via efficiently metabolizing intestinal aa. To identify the responsible microbes/genes, we developed a metabolomics-based assay to screen 104 commensals and identified candidates that efficiently utilize aa. Using genetics, we identified multiple responsible metabolic genes in phylogenetically diverse microbes. By colonizing germ-free mice with the wild-type strain and their isogenic mutant deficient in individual aa-metabolizing genes, we found that these genes regulate the availability of gut and circulatory aa. Notably, microbiota genes for branched-chain amino acids (BCAAs) and tryptophan metabolism indirectly affect host glucose homeostasis via peripheral serotonin. Collectively, at single-gene level, this work characterizes a microbiota-encoded metabolic activity that affects host nutrient homeostasis and provides a roadmap to interrogate microbiota-dependent activity to improve human health.


Asunto(s)
Aminoácidos de Cadena Ramificada , Aminoácidos , Microbioma Gastrointestinal , Homeostasis , Triptófano , Animales , Microbioma Gastrointestinal/fisiología , Ratones , Aminoácidos/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Triptófano/metabolismo , Ratones Endogámicos C57BL , Nutrientes/metabolismo , Intestinos/microbiología , Humanos , Metabolómica , Glucosa/metabolismo , Serotonina/metabolismo , Vida Libre de Gérmenes , Bacterias/metabolismo , Bacterias/genética , Bacterias/clasificación , Masculino
19.
Microbiome ; 12(1): 86, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730492

RESUMEN

BACKGROUND: Parasitic helminths influence the composition of the gut microbiome. However, the microbiomes of individuals living in helminth-endemic regions are understudied. The Orang Asli, an indigenous population in Malaysia with high burdens of the helminth Trichuris trichiura, display microbiotas enriched in Clostridiales, an order of spore-forming obligate anaerobes with immunogenic properties. We previously isolated novel Clostridiales that were enriched in these individuals and found that a subset promoted the Trichuris life cycle. In this study, we aimed to further characterize the functional properties of these bacteria. RESULTS: Clostridiales isolates were profiled for their ability to perform 57 enzymatic reactions and produce short-chain fatty acids (SCFAs) and hydrogen sulfide, revealing that these bacteria were capable of a range of activities associated with metabolism and host response. Consistent with this finding, monocolonization of mice with individual isolates identified bacteria that were potent inducers of regulatory T-cell (Treg) differentiation in the colon. Comparisons between variables revealed by these studies identified enzymatic properties correlated with Treg induction and Trichuris egg hatching. CONCLUSION: We identified Clostridiales species that are sufficient to induce high levels of Tregs. We also identified a set of metabolic activities linked with Treg differentiation and Trichuris egg hatching mediated by these newly isolated bacteria. Altogether, this study provides functional insights into the microbiotas of individuals residing in a helminth-endemic region. Video Abstract.


Asunto(s)
Diferenciación Celular , Clostridiales , Microbioma Gastrointestinal , Linfocitos T Reguladores , Trichuris , Animales , Linfocitos T Reguladores/inmunología , Ratones , Malasia , Clostridiales/aislamiento & purificación , Humanos , Ácidos Grasos Volátiles/metabolismo , Femenino , Tricuriasis/parasitología , Tricuriasis/inmunología , Tricuriasis/microbiología
20.
J Exp Med ; 221(5)2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38506708

RESUMEN

Innate lymphoid cells (ILCs) can promote host defense, chronic inflammation, or tissue protection and are regulated by cytokines and neuropeptides. However, their regulation by diet and microbiota-derived signals remains unclear. We show that an inulin fiber diet promotes Tph1-expressing inflammatory ILC2s (ILC2INFLAM) in the colon, which produce IL-5 but not tissue-protective amphiregulin (AREG), resulting in the accumulation of eosinophils. This exacerbates inflammation in a murine model of intestinal damage and inflammation in an ILC2- and eosinophil-dependent manner. Mechanistically, the inulin fiber diet elevated microbiota-derived bile acids, including cholic acid (CA) that induced expression of ILC2-activating IL-33. In IBD patients, bile acids, their receptor farnesoid X receptor (FXR), IL-33, and eosinophils were all upregulated compared with controls, implicating this diet-microbiota-ILC2 axis in human IBD pathogenesis. Together, these data reveal that dietary fiber-induced changes in microbial metabolites operate as a rheostat that governs protective versus pathologic ILC2 responses with relevance to precision nutrition for inflammatory diseases.


Asunto(s)
Inmunidad Innata , Enfermedades Inflamatorias del Intestino , Humanos , Animales , Ratones , Interleucina-33 , Inulina , Linfocitos , Fibras de la Dieta , Ácidos y Sales Biliares , Inflamación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA