Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell ; 185(3): 513-529.e21, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35120663

RESUMEN

The human gut microbiota resides within a diverse chemical environment challenging our ability to understand the forces shaping this ecosystem. Here, we reveal that fitness of the Bacteroidales, the dominant order of bacteria in the human gut, is an emergent property of glycans and one specific metabolite, butyrate. Distinct sugars serve as strain-variable fitness switches activating context-dependent inhibitory functions of butyrate. Differential fitness effects of butyrate within the Bacteroides are mediated by species-level variation in Acyl-CoA thioesterase activity and nucleotide polymorphisms regulating an Acyl-CoA transferase. Using in vivo multi-omic profiles, we demonstrate Bacteroides fitness in the human gut is associated together, but not independently, with Acyl-CoA transferase expression and butyrate. Our data reveal that each strain of the Bacteroides exists within a unique fitness landscape based on the interaction of chemical components unpredictable by the effect of each part alone mediated by flexibility in the core genome.


Asunto(s)
Microbioma Gastrointestinal , Metaboloma , Polisacáridos/metabolismo , Acilcoenzima A/metabolismo , Secuencia de Aminoácidos , Aminoácidos de Cadena Ramificada/metabolismo , Bacteroidetes/efectos de los fármacos , Bacteroidetes/genética , Bacteroidetes/crecimiento & desarrollo , Butiratos/química , Butiratos/farmacología , Coenzima A Transferasas/química , Coenzima A Transferasas/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Variación Genética/efectos de los fármacos , Concentración de Iones de Hidrógeno , Metaboloma/efectos de los fármacos , Metaboloma/genética , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Especificidad de la Especie , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética , Transcripción Genética/efectos de los fármacos
2.
Nature ; 591(7851): 633-638, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33627867

RESUMEN

The gut microbiota of preterm infants develops predictably1-7, with pioneer species colonizing the gut after birth, followed by an ordered succession of microorganisms. The gut microbiota is vital to the health of preterm infants8,9, but the forces that shape these predictable dynamics of microbiome assembly are unknown. The environment, the host and interactions between microorganisms all potentially shape the dynamics of the microbiota, but in such a complex ecosystem, identifying the specific role of any individual factor is challenging10-14. Here we use multi-kingdom absolute abundance quantification, ecological modelling and experimental validation to address this challenge. We quantify the absolute dynamics of bacteria, fungi and archaea in a longitudinal cohort of 178 preterm infants. We uncover microbial blooms and extinctions, and show that there is an inverse correlation between bacterial and fungal loads in the infant gut. We infer computationally and demonstrate experimentally in vitro and in vivo that predictable assembly dynamics may be driven by directed, context-dependent interactions between specific microorganisms. Mirroring the dynamics of macroscopic ecosystems15-17, a late-arriving member of the microbiome, Klebsiella, exploits the pioneer microorganism, Staphylococcus, to gain a foothold within the gut. Notably, we find that interactions between different kingdoms can influence assembly, with a single fungal species-Candida albicans-inhibiting multiple dominant genera of gut bacteria. Our work reveals the centrality of simple microbe-microbe interactions in shaping host-associated microbiota, which is critical both for our understanding of microbiota ecology and for targeted microbiota interventions.


Asunto(s)
Biodiversidad , Microbioma Gastrointestinal , Recien Nacido Prematuro , Carga Bacteriana , Dieta , Femenino , Edad Gestacional , Humanos , Lactante , Recién Nacido , Masculino , Interacciones Microbianas , Reproducibilidad de los Resultados
3.
PLoS Biol ; 19(2): e3001116, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33606675

RESUMEN

Humans and many other hosts establish a diverse community of beneficial microbes anew each generation. The order and identity of incoming symbionts is critical for health, but what determines the success of the assembly process remains poorly understood. Here we develop ecological theory to identify factors important for microbial community assembly. Our method maps out all feasible pathways for the assembly of a given microbiome-with analogies to the mutational maps underlying fitness landscapes in evolutionary biology. Building these "assembly maps" reveals a tradeoff at the heart of the assembly process. Ecological dependencies between members of the microbiota make assembly predictable-and can provide metabolic benefits to the host-but these dependencies may also create barriers to assembly. This effect occurs because interdependent species can fail to establish when each relies on the other to colonize first. We support our predictions with published data from the assembly of the preterm infant microbiota, where we find that ecological dependence is associated with a predictable order of arrival. Our models also suggest that hosts can overcome barriers to assembly via mechanisms that either promote the uptake of multiple symbiont species in one step or feed early colonizers. This predicted importance of host feeding is supported by published data on the impacts of breast milk in the assembly of the human microbiome. We conclude that both microbe to microbe and host to microbe interactions are important for the trajectory of microbiome assembly.


Asunto(s)
Recien Nacido Prematuro , Microbiota , Humanos , Recién Nacido , Leche Humana/microbiología , Modelos Teóricos , Simbiosis
4.
Nucleic Acids Res ; 47(8): 4292-4307, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30820540

RESUMEN

Bacterial small regulatory RNAs (sRNAs) are the most abundant class of post-transcriptional regulators and have been well studied in Gram-negative bacteria. Little is known about the functions and mechanisms of sRNAs in high GC Gram-positive bacteria including Mycobacterium and Streptomyces. Here, we performed an in-depth study of 6C sRNA of Mycobacterium tuberculosis, which is conserved among high GC Gram-positive bacteria. Forty-seven genes were identified as possible direct targets of 6C sRNA and 15 of them were validated using an in vivo translational lacZ fusion system. We found that 6C sRNA plays a pleotropic role and regulates genes involved in various cellular processes, including DNA replication and protein secretion. Mapping the interactions of 6C sRNA with mRNA targets panD and dnaB revealed that the C-rich loops of 6C sRNA act as direct binding sites to mRNA targets. Unlike in Gram-negative bacteria where RNA binding proteins Hfq and ProQ are required, the interactions of 6C sRNA with mRNAs appear to be independent of RNA chaperones. Our findings suggest that the multiple G-C pairings between single stranded regions are sufficient to establish stable interactions between 6C sRNA and mRNA targets, providing a mechanism for sRNAs in high GC Gram-positive bacteria.


Asunto(s)
Proteínas Bacterianas/genética , Carboxiliasas/genética , AdnB Helicasas/genética , Regulación Bacteriana de la Expresión Génica , Mycobacterium tuberculosis/genética , ARN Mensajero/genética , ARN Pequeño no Traducido/genética , Proteínas Bacterianas/metabolismo , Emparejamiento Base , Secuencia de Bases , Carboxiliasas/metabolismo , Replicación del ADN , AdnB Helicasas/metabolismo , Operón Lac , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mycobacterium tuberculosis/metabolismo , Conformación de Ácido Nucleico , Biosíntesis de Proteínas , Transporte de Proteínas , ARN Mensajero/metabolismo , ARN Pequeño no Traducido/metabolismo
5.
RNA ; 23(10): 1525-1538, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28724535

RESUMEN

CRISPR-Cas is a bacterial and archaeal adaptive immune system that uses short, invader-derived sequences termed spacers to target invasive nucleic acids. Upon recognition of previously encountered invaders, the system can stimulate secondary spacer acquisitions, a process known as primed adaptation. Previous studies of primed adaptation have been complicated by intrinsically high interference efficiency of most systems against bona fide targets. As such, most primed adaptation to date has been studied within the context of imperfect sequence complementarity between spacers and targets. Here, we take advantage of a native type I-C CRISPR-Cas system in Legionella pneumophila that displays robust primed adaptation even within the context of a perfectly matched target. Using next-generation sequencing to survey acquired spacers, we observe strand bias and positional preference that are consistent with a 3'-5' translocation of the adaptation machinery. We show that spacer acquisition happens in a wide range of frequencies across the plasmid, including a remarkable hotspot that predominates irrespective of the priming strand. We systematically characterize protospacer sequence constraints in both adaptation and interference and reveal extensive flexibilities regarding the protospacer adjacent motif in both processes. Lastly, in a strain with a genetically truncated CRISPR array, we observe increased interference efficiency, which, when coupled with forced maintenance of a targeted plasmid, provides a useful experimental system to study spacer loss. Based on these observations, we propose that the Legionella pneumophila type I-C system represents a powerful model to study primed adaptation and the interplay between CRISPR interference and adaptation.


Asunto(s)
Sistemas CRISPR-Cas , Legionella pneumophila/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Motivos de Nucleótidos , Plásmidos
6.
Mol Syst Biol ; 12(12): 893, 2016 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-27986836

RESUMEN

Pathogens deliver complex arsenals of translocated effector proteins to host cells during infection, but the extent to which these proteins are regulated once inside the eukaryotic cell remains poorly defined. Among all bacterial pathogens, Legionella pneumophila maintains the largest known set of translocated substrates, delivering over 300 proteins to the host cell via its Type IVB, Icm/Dot translocation system. Backed by a few notable examples of effector-effector regulation in L. pneumophila, we sought to define the extent of this phenomenon through a systematic analysis of effector-effector functional interaction. We used Saccharomyces cerevisiae, an established proxy for the eukaryotic host, to query > 108,000 pairwise genetic interactions between two compatible expression libraries of ~330 L. pneumophila-translocated substrates. While capturing all known examples of effector-effector suppression, we identify fourteen novel translocated substrates that suppress the activity of other bacterial effectors and one pair with synergistic activities. In at least nine instances, this regulation is direct-a hallmark of an emerging class of proteins called metaeffectors, or "effectors of effectors". Through detailed structural and functional analysis, we show that metaeffector activity derives from a diverse range of mechanisms, shapes evolution, and can be used to reveal important aspects of each cognate effector's function. Metaeffectors, along with other, indirect, forms of effector-effector modulation, may be a common feature of many intracellular pathogens-with unrealized potential to inform our understanding of how pathogens regulate their interactions with the host cell.


Asunto(s)
Proteínas Bacterianas/metabolismo , Legionella pneumophila/patogenicidad , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Legionella pneumophila/metabolismo , Modelos Biológicos , Mapas de Interacción de Proteínas , Biología de Sistemas/métodos
7.
Cell Microbiol ; 18(10): 1319-38, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26936325

RESUMEN

Clustered regularly interspaced short palindromic repeats with CRISPR-associated gene (CRISPR-Cas) systems are widely recognized as critical genome defense systems that protect microbes from external threats such as bacteriophage infection. Several isolates of the intracellular pathogen Legionella pneumophila possess multiple CRISPR-Cas systems (type I-C, type I-F and type II-B), yet the targets of these systems remain unknown. With the recent observation that at least one of these systems (II-B) plays a non-canonical role in supporting intracellular replication, the possibility remained that these systems are vestigial genome defense systems co-opted for other purposes. Our data indicate that this is not the case. Using an established plasmid transformation assay, we demonstrate that type I-C, I-F and II-B CRISPR-Cas provide protection against spacer targets. We observe efficient laboratory acquisition of new spacers under 'priming' conditions, in which initially incomplete target elimination leads to the generation of new spacers and ultimate loss of the invasive DNA. Critically, we identify the first known target of L. pneumophila CRISPR-Cas: a 30 kb episome of unknown function whose interbacterial transfer is guarded against by CRISPR-Cas. We provide evidence that the element can subvert CRISPR-Cas by mutating its targeted sequences - but that primed spacer acquisition may limit this mechanism of escape. Rather than generally impinging on bacterial fitness, this element drives a host specialization event - with improved fitness in Acanthamoeba but a reduced ability to replicate in other hosts and conditions. These observations add to a growing body of evidence that host range restriction can serve as an existential threat to L. pneumophila in the wild.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Legionella pneumophila/genética , Acanthamoeba castellanii/microbiología , Secuencia de Bases , Secuencia Conservada , Evolución Molecular , Genes Bacterianos , Interacciones Huésped-Patógeno , Legionella pneumophila/crecimiento & desarrollo , Viabilidad Microbiana , Análisis de Secuencia de ADN
8.
PLoS Pathog ; 10(11): e1004500, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25375226

RESUMEN

The bacterial H-NS protein silences expression from sequences with higher AT-content than the host genome and is believed to buffer the fitness consequences associated with foreign gene acquisition. Loss of H-NS results in severe growth defects in Salmonella, but the underlying reasons were unclear. An experimental evolution approach was employed to determine which secondary mutations could compensate for the loss of H-NS in Salmonella. Six independently derived S. Typhimurium hns mutant strains were serially passaged for 300 generations prior to whole genome sequencing. Growth rates of all lineages dramatically improved during the course of the experiment. Each of the hns mutant lineages acquired missense mutations in the gene encoding the H-NS paralog StpA encoding a poorly understood H-NS paralog, while 5 of the mutant lineages acquired deletions in the genes encoding the Salmonella Pathogenicity Island-1 (SPI-1) Type 3 secretion system critical to invoke inflammation. We further demonstrate that SPI-1 misregulation is a primary contributor to the decreased fitness in Salmonella hns mutants. Three of the lineages acquired additional loss of function mutations in the PhoPQ virulence regulatory system. Similarly passaged wild type Salmonella lineages did not acquire these mutations. The stpA missense mutations arose in the oligomerization domain and generated proteins that could compensate for the loss of H-NS to varying degrees. StpA variants most able to functionally substitute for H-NS displayed altered DNA binding and oligomerization properties that resembled those of H-NS. These findings indicate that H-NS was central to the evolution of the Salmonellae by buffering the negative fitness consequences caused by the secretion system that is the defining characteristic of the species.


Asunto(s)
Proteínas Bacterianas , Proteínas de Unión al ADN , Evolución Molecular , Regulación Bacteriana de la Expresión Génica/fisiología , Silenciador del Gen/fisiología , Islas Genómicas/fisiología , Salmonella , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Mutación , Salmonella/genética , Salmonella/metabolismo
9.
Appl Environ Microbiol ; 80(4): 1441-54, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24334670

RESUMEN

Although only partially understood, multicellular behavior is relatively common in bacterial pathogens. Bacterial aggregates can resist various host defenses and colonize their environment more efficiently than planktonic cells. For the waterborne pathogen Legionella pneumophila, little is known about the roles of autoaggregation or the parameters which allow cell-cell interactions to occur. Here, we determined the endogenous and exogenous factors sufficient to allow autoaggregation to take place in L. pneumophila. We show that isolates from Legionella species which do not produce the Legionella collagen-like protein (Lcl) are deficient in autoaggregation. Targeted deletion of the Lcl-encoding gene (lpg2644) and the addition of Lcl ligands impair the autoaggregation of L. pneumophila. In addition, Lcl-induced autoaggregation requires divalent cations. Escherichia coli producing surface-exposed Lcl is able to autoaggregate and shows increased biofilm production. We also demonstrate that L. pneumophila infection of Acanthamoeba castellanii and Hartmanella vermiformis is potentiated under conditions which promote Lcl dependent autoaggregation. Overall, this study shows that L. pneumophila is capable of autoaggregating in a process that is mediated by Lcl in a divalent-cation-dependent manner. It also reveals that Lcl potentiates the ability of L. pneumophila to come in contact, attach, and infect amoebae.


Asunto(s)
Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Legionella pneumophila/fisiología , Fagocitos/microbiología , Acanthamoeba castellanii/microbiología , Proteínas Bacterianas/genética , Cationes Bivalentes/metabolismo , Escherichia coli/genética , Escherichia coli/fisiología , Eliminación de Gen , Legionella pneumophila/genética , Lobosea/microbiología
10.
EMBO Rep ; 13(4): 371-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22402663

RESUMEN

FOXO transcription factors have a critical role in oxidative stress-induced neuronal cell death. A variety of post-translational modifications of FOXO family proteins have been reported, including phosphorylation, acetylation, ubiqutination and recently arginine methylation. Here, we demonstrate that the methyltransferase Set9 methylates FOXO3 at lysine 270. Methylation of FOXO3 leads to the inhibition of its DNA-binding activity and transactivation. Accordingly, lysine methylation reduces oxidative stress-induced and FOXO3-mediated Bim expression and neuronal apoptosis in neurons. Collectively, these findings define a novel modification of FOXO3 and show that lysine methylation negatively regulates FOXO3-mediated transcription and neuronal apoptosis.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Lisina/metabolismo , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo , Secuencia de Aminoácidos , Animales , Muerte Celular , ADN/metabolismo , Proteína Forkhead Box O3 , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Metilación , Ratones , Datos de Secuencia Molecular , Unión Proteica , Transcripción Genética
11.
J Clin Invest ; 131(3)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33529166

RESUMEN

The coat protein I (COPI) complex mediates retrograde trafficking from the Golgi to the endoplasmic reticulum (ER). Five siblings with persistent bacterial and viral infections and defective humoral and cellular immunity had a homozygous p.K652E mutation in the γ1 subunit of COPI (γ1-COP). The mutation disrupts COPI binding to the KDEL receptor and impairs the retrieval of KDEL-bearing chaperones from the Golgi to the ER. Homozygous Copg1K652E mice had increased ER stress in activated T and B cells, poor antibody responses, and normal numbers of T cells that proliferated normally, but underwent increased apoptosis upon activation. Exposure of the mutants to pet store mice caused weight loss, lymphopenia, and defective T cell proliferation that recapitulated the findings in the patients. The ER stress-relieving agent tauroursodeoxycholic acid corrected the immune defects of the mutants and reversed the phenotype they acquired following exposure to pet store mice. This study establishes the role of γ1-COP in the ER retrieval of KDEL-bearing chaperones and thereby the importance of ER homeostasis in adaptive immunity.


Asunto(s)
Apoptosis/inmunología , Linfocitos B/inmunología , Estrés del Retículo Endoplásmico/inmunología , Activación de Linfocitos , Mutación Missense , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/inmunología , Sustitución de Aminoácidos , Animales , Apoptosis/genética , Proteína Coatómero/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Estrés del Retículo Endoplásmico/genética , Aparato de Golgi/genética , Aparato de Golgi/inmunología , Humanos , Ratones , Ratones Mutantes , Receptores de Péptidos/genética , Receptores de Péptidos/inmunología , Inmunodeficiencia Combinada Grave/genética
12.
Inflamm Bowel Dis ; 22(1): 1-12, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26444104

RESUMEN

BACKGROUND: Mucosal-associated Escherichia coli are commonly found in inflamed tissues during inflammatory bowel disease (IBD). These bacteria often possess an adherent and invasive phenotype but lack virulence-associated features of well-described intestinal E. coli pathogens, and are of diverse serology and phylotypes, making it difficult to correlate strain characteristics with exacerbations of disease. METHODS: The genome sequences of 14 phenotypically assigned adherent-invasive Escherichia coli (AIEC) isolates obtained from intestinal biopsies of patients with IBD were compared with the genome sequences of 37 other pathogenic and commensal E. coli available from public databases. RESULTS: Core genome-based phylogenetic analyses and genome-wide comparison of genetic content established the existence of a closely related cluster of AIEC strains with 3 distinct genetic insertions differentiating them from commensal E. coli. These strains are of the B2 phylotype have a variant type VI secretion system (T6SS-1), and are highly related to extraintestinal pathogenic E. coli, suggesting that these 2 clinically distinct pathovars have common virulence strategies. Four other mucosally adherent E. coli strains from patients with IBD were of diverse phylogenetic origins and lacked the 3 genetic features, suggesting that they are not related to the B2 AIEC cluster. Although AIEC are often considered as having a unique association with Crohn's disease, isolates from Crohn's disease and ulcerative colitis were genetically indistinguishable. CONCLUSIONS: B2 AIEC thus represent a closely related cluster of IBD-associated E. coli strains that are distinct from normal commensal isolates, and which should be considered separately from the phenotypically similar but genetically distinct non-B2 AIEC strains when considering their association with intestinal pathogenesis.


Asunto(s)
Adhesión Bacteriana/genética , Infecciones por Escherichia coli/microbiología , Escherichia coli/genética , Escherichia coli/patogenicidad , Variación Genética/genética , Genoma Bacteriano , Enfermedades Inflamatorias del Intestino/microbiología , Escherichia coli/clasificación , Infecciones por Escherichia coli/diagnóstico , Humanos , Enfermedades Inflamatorias del Intestino/diagnóstico , Fenotipo , Filogenia , Virulencia
13.
PLoS One ; 8(5): e64129, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23717549

RESUMEN

Over 20 years ago, two groups independently domesticated Legionella pneumophila from a clinical isolate of bacteria collected during the first recognized outbreak of Legionnaires' disease (at the 1976 American Legion's convention in Philadelphia). These two laboratory strains, JR32 and Lp01, along with their derivatives, have been disseminated to a number of laboratories around the world and form the cornerstone of much of the research conducted on this important pathogen to date. Nevertheless, no exhaustive examination of the genetic distance between these strains and their clinical progenitor has been performed thus far. Such information is of paramount importance for making sense of several phenotypic differences observed between these strains. As environmental replication of L. pneumophila is thought to exclusively occur within natural protozoan hosts, retrospective analysis of the domestication and axenic culture of the Philadelphia-1 progenitor strain by two independent groups also provides an excellent opportunity to uncover evidence of adaptation to the laboratory environment. To reconstruct the phylogenetic relationships between the common laboratory strains of L. pneumophila Philadelphia-1 and their clinical ancestor, we performed whole-genome Illumina resequencing of the two founders of each laboratory lineage: JR32 and Lp01. As expected from earlier, targeted studies, Lp01 and JR32 contain large deletions in the lvh and tra regions, respectively. By sequencing additional strains derived from Lp01 (Lp02 and Lp03), we retraced the phylogeny of these strains relative to their reported ancestor, thereby reconstructing the evolutionary dynamics of each laboratory lineage from genomic data.


Asunto(s)
Genoma Bacteriano , Legionella pneumophila/clasificación , Filogenia , Legionella pneumophila/genética , Mutación , Polimorfismo Genético
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda