Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.403
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 31: 73-106, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23215645

RESUMEN

Inflammasomes are cytosolic multiprotein complexes that assemble in response to a variety of infectious and noxious insults. Inflammasomes play a critical role in the initiation of innate immune responses, primarily by serving as platforms for the activation of inflammatory caspase proteases. One such caspase, CASPASE-1 (CASP1), initiates innate immune responses by cleaving pro-IL-1ß and pro-IL-18, leading to their activation and release. CASP1 and another inflammatory caspase termed CASP11 can also initiate a rapid and inflammatory form of cell death termed pyroptosis. Several distinct inflammasomes have been described, each of which contains a unique sensor protein of the NLR (nucleotide-binding domain, leucine-rich repeat-containing) superfamily or the PYHIN (PYRIN and HIN-200 domain-containing) superfamily. Here we describe the surprisingly diverse mechanisms by which NLR/PYHIN proteins sense bacteria and initiate innate immune responses. We conclude that inflammasomes represent a highly adaptable scaffold ideally suited for detecting and initiating rapid innate responses to diverse and rapidly evolving bacteria.


Asunto(s)
Bacterias/patogenicidad , Inflamasomas/metabolismo , Animales , Bacillus anthracis/patogenicidad , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas Adaptadoras de Señalización CARD/fisiología , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/fisiología , Flagelos/metabolismo , Flagelos/fisiología , Humanos , Inflamasomas/genética , Inflamasomas/fisiología , Legionella pneumophila/patogenicidad , Listeria monocytogenes/patogenicidad , Salmonella typhimurium/patogenicidad
2.
Cell ; 179(3): 703-712.e7, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31587897

RESUMEN

Peptidoglycan (PG) is a defining feature of bacteria, involved in cell division, shape, and integrity. We previously reported that several genes related to PG biosynthesis were horizontally transferred from bacteria to the nuclear genome of mealybugs. Mealybugs are notable for containing a nested bacteria-within-bacterium endosymbiotic structure in specialized insect cells, where one bacterium, Moranella, lives in the cytoplasm of another bacterium, Tremblaya. Here we show that horizontally transferred genes on the mealybug genome work together with genes retained on the Moranella genome to produce a PG layer exclusively at the Moranella cell periphery. Furthermore, we show that an insect protein encoded by a horizontally transferred gene of bacterial origin is transported into the Moranella cytoplasm. These results provide a striking parallel to the genetic and biochemical mosaicism found in organelles, and prove that multiple horizontally transferred genes can become integrated into a functional pathway distributed between animal and bacterial endosymbiont genomes.


Asunto(s)
Bacterias/genética , Transferencia de Gen Horizontal , Hemípteros/genética , Peptidoglicano/biosíntesis , Simbiosis , Animales , Bacterias/patogenicidad , Genes Bacterianos , Hemípteros/microbiología , Interacciones Huésped-Patógeno , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Peptidoglicano/genética
3.
Nat Immunol ; 22(2): 216-228, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33462454

RESUMEN

CD4+ effector lymphocytes (Teff) are traditionally classified by the cytokines they produce. To determine the states that Teff cells actually adopt in frontline tissues in vivo, we applied single-cell transcriptome and chromatin analyses to colonic Teff cells in germ-free or conventional mice or in mice after challenge with a range of phenotypically biasing microbes. Unexpected subsets were marked by the expression of the interferon (IFN) signature or myeloid-specific transcripts, but transcriptome or chromatin structure could not resolve discrete clusters fitting classic helper T cell (TH) subsets. At baseline or at different times of infection, transcripts encoding cytokines or proteins commonly used as TH markers were distributed in a polarized continuum, which was functionally validated. Clones derived from single progenitors gave rise to both IFN-γ- and interleukin (IL)-17-producing cells. Most of the transcriptional variance was tied to the infecting agent, independent of the cytokines produced, and chromatin variance primarily reflected activities of activator protein (AP)-1 and IFN-regulatory factor (IRF) transcription factor (TF) families, not the canonical subset master regulators T-bet, GATA3 or RORγ.


Asunto(s)
Bacterias/patogenicidad , Infecciones Bacterianas/microbiología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/parasitología , Colon/microbiología , Colon/parasitología , Microbioma Gastrointestinal , Heligmosomatoidea/patogenicidad , Parasitosis Intestinales/parasitología , Animales , Bacterias/inmunología , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Cromatina/genética , Cromatina/metabolismo , Citrobacter rodentium/inmunología , Citrobacter rodentium/patogenicidad , Colon/inmunología , Colon/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Heligmosomatoidea/inmunología , Interacciones Huésped-Patógeno , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Parasitosis Intestinales/genética , Parasitosis Intestinales/inmunología , Parasitosis Intestinales/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Nematospiroides dubius/inmunología , Nematospiroides dubius/patogenicidad , Nippostrongylus/inmunología , Nippostrongylus/patogenicidad , Fenotipo , Salmonella enterica/inmunología , Salmonella enterica/patogenicidad , Análisis de la Célula Individual , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Transcriptoma
4.
Cell ; 175(4): 973-983.e14, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388454

RESUMEN

Roots of healthy plants are inhabited by soil-derived bacteria, fungi, and oomycetes that have evolved independently in distinct kingdoms of life. How these microorganisms interact and to what extent those interactions affect plant health are poorly understood. We examined root-associated microbial communities from three Arabidopsis thaliana populations and detected mostly negative correlations between bacteria and filamentous microbial eukaryotes. We established microbial culture collections for reconstitution experiments using germ-free A. thaliana. In plants inoculated with mono- or multi-kingdom synthetic microbial consortia, we observed a profound impact of the bacterial root microbiota on fungal and oomycetal community structure and diversity. We demonstrate that the bacterial microbiota is essential for plant survival and protection against root-derived filamentous eukaryotes. Deconvolution of 2,862 binary bacterial-fungal interactions ex situ, combined with community perturbation experiments in planta, indicate that biocontrol activity of bacterial root commensals is a redundant trait that maintains microbial interkingdom balance for plant health.


Asunto(s)
Arabidopsis/microbiología , Consorcios Microbianos , Raíces de Plantas/microbiología , Arabidopsis/fisiología , Bacterias/patogenicidad , Hongos/patogenicidad , Simbiosis
5.
Annu Rev Cell Dev Biol ; 34: 357-379, 2018 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-30095291

RESUMEN

Microbial nucleic acids are major signatures of invading pathogens, and their recognition by various host pattern recognition receptors (PRRs) represents the first step toward an efficient innate immune response to clear the pathogens. The nucleic acid-sensing PRRs are localized at the plasma membrane, the cytosol, and/or various cellular organelles. Sensing of nucleic acids and signaling by PRRs involve recruitment of distinct signaling components, and PRRs are intensively regulated by cellular organelle trafficking. PRR-mediated innate immune responses are also heavily regulated by posttranslational modifications, including phosphorylation, polyubiquitination, sumoylation, and glutamylation. In this review, we focus on our current understanding of recognition of microbial nucleic acid by PRRs, particularly on their regulation by organelle trafficking and posttranslational modifications. We also discuss how sensing of self nucleic acids and dysregulation of PRR-mediated signaling lead to serious human diseases.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Inmunidad Innata/genética , Ácidos Nucleicos/genética , Receptores de Reconocimiento de Patrones/genética , Bacterias/genética , Bacterias/patogenicidad , Citoplasma/inmunología , Citoplasma/microbiología , ADN Bacteriano/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Ácidos Nucleicos/inmunología , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Transducción de Señal/genética
6.
Nature ; 628(8006): 171-179, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38509360

RESUMEN

The myriad microorganisms that live in close association with humans have diverse effects on physiology, yet the molecular bases for these impacts remain mostly unknown1-3. Classical pathogens often invade host tissues and modulate immune responses through interactions with human extracellular and secreted proteins (the 'exoproteome'). Commensal microorganisms may also facilitate niche colonization and shape host biology by engaging host exoproteins; however, direct exoproteome-microbiota interactions remain largely unexplored. Here we developed and validated a novel technology, BASEHIT, that enables proteome-scale assessment of human exoproteome-microbiome interactions. Using BASEHIT, we interrogated more than 1.7 million potential interactions between 519 human-associated bacterial strains from diverse phylogenies and tissues of origin and 3,324 human exoproteins. The resulting interactome revealed an extensive network of transkingdom connectivity consisting of thousands of previously undescribed host-microorganism interactions involving 383 strains and 651 host proteins. Specific binding patterns within this network implied underlying biological logic; for example, conspecific strains exhibited shared exoprotein-binding patterns, and individual tissue isolates uniquely bound tissue-specific exoproteins. Furthermore, we observed dozens of unique and often strain-specific interactions with potential roles in niche colonization, tissue remodelling and immunomodulation, and found that strains with differing host interaction profiles had divergent interactions with host cells in vitro and effects on the host immune system in vivo. Overall, these studies expose a previously unexplored landscape of molecular-level host-microbiota interactions that may underlie causal effects of indigenous microorganisms on human health and disease.


Asunto(s)
Bacterias , Interacciones Microbiota-Huesped , Microbiota , Filogenia , Proteoma , Simbiosis , Animales , Femenino , Humanos , Ratones , Bacterias/clasificación , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/patogenicidad , Interacciones Microbiota-Huesped/inmunología , Interacciones Microbiota-Huesped/fisiología , Tropismo al Anfitrión , Microbiota/inmunología , Microbiota/fisiología , Especificidad de Órganos , Unión Proteica , Proteoma/inmunología , Proteoma/metabolismo , Reproducibilidad de los Resultados
7.
Nature ; 617(7962): 807-817, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37198490

RESUMEN

Microbial organisms have key roles in numerous physiological processes in the human body and have recently been shown to modify the response to immune checkpoint inhibitors1,2. Here we aim to address the role of microbial organisms and their potential role in immune reactivity against glioblastoma. We demonstrate that HLA molecules of both glioblastoma tissues and tumour cell lines present bacteria-specific peptides. This finding prompted us to examine whether tumour-infiltrating lymphocytes (TILs) recognize tumour-derived bacterial peptides. Bacterial peptides eluted from HLA class II molecules are recognized by TILs, albeit very weakly. Using an unbiased antigen discovery approach to probe the specificity of a TIL CD4+ T cell clone, we show that it recognizes a broad spectrum of peptides from pathogenic bacteria, commensal gut microbiota and also glioblastoma-related tumour antigens. These peptides were also strongly stimulatory for bulk TILs and peripheral blood memory cells, which then respond to tumour-derived target peptides. Our data hint at how bacterial pathogens and bacterial gut microbiota can be involved in specific immune recognition of tumour antigens. The unbiased identification of microbial target antigens for TILs holds promise for future personalized tumour vaccination approaches.


Asunto(s)
Antígenos de Neoplasias , Bacterias , Proteínas Bacterianas , Glioblastoma , Linfocitos Infiltrantes de Tumor , Fragmentos de Péptidos , Humanos , Antígenos de Neoplasias/inmunología , Proteínas Bacterianas/inmunología , Vacunas contra el Cáncer/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Microbioma Gastrointestinal/inmunología , Glioblastoma/inmunología , Glioblastoma/patología , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos HLA/inmunología , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Fragmentos de Péptidos/inmunología , Simbiosis , Bacterias/inmunología , Bacterias/patogenicidad
8.
Nature ; 607(7919): 563-570, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35831502

RESUMEN

Gut commensal bacteria with the ability to translocate across the intestinal barrier can drive the development of diverse immune-mediated diseases1-4. However, the key factors that dictate bacterial translocation remain unclear. Recent studies have revealed that gut microbiota strains can adapt and evolve throughout the lifetime of the host5-9, raising the possibility that changes in individual commensal bacteria themselves over time may affect their propensity to elicit inflammatory disease. Here we show that within-host evolution of the model gut pathobiont Enterococcus gallinarum facilitates bacterial translocation and initiation of inflammation. Using a combination of in vivo experimental evolution and comparative genomics, we found that E. gallinarum diverges into independent lineages adapted to colonize either luminal or mucosal niches in the gut. Compared with ancestral and luminal E. gallinarum, mucosally adapted strains evade detection and clearance by the immune system, exhibit increased translocation to and survival within the mesenteric lymph nodes and liver, and induce increased intestinal and hepatic inflammation. Mechanistically, these changes in bacterial behaviour are associated with non-synonymous mutations or insertion-deletions in defined regulatory genes in E. gallinarum, altered microbial gene expression programs and remodelled cell wall structures. Lactobacillus reuteri also exhibited broadly similar patterns of divergent evolution and enhanced immune evasion in a monocolonization-based model of within-host evolution. Overall, these studies define within-host evolution as a critical regulator of commensal pathogenicity that provides a unique source of stochasticity in the development and progression of microbiota-driven disease.


Asunto(s)
Bacterias , Traslocación Bacteriana , Evolución Biológica , Microbioma Gastrointestinal , Hígado , Bacterias/genética , Bacterias/inmunología , Bacterias/patogenicidad , Traslocación Bacteriana/genética , Pared Celular/genética , Enterococcus/genética , Enterococcus/inmunología , Microbioma Gastrointestinal/genética , Genómica , Interacciones Huésped-Patógeno/inmunología , Humanos , Inflamación/microbiología , Inflamación/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/inmunología , Hígado/microbiología , Hígado/patología , Ganglios Linfáticos/microbiología , Mutación , Procesos Estocásticos , Simbiosis/genética , Simbiosis/inmunología
9.
Mol Cell ; 78(4): 584-596, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32234490

RESUMEN

Many genomic studies have revealed associations between the gut microbiota composition and host metabolism. These observations led to the idea that a causal relationship could exist between the microbiota and metabolic diseases, a concept supported by studies showing compositional changes in the microbial community in metabolic diseases and transmissibility of host phenotype via microbiota transfer. Accumulating data suggest that the microbiota may affect host metabolic phenotypes through the production of metabolites. These bioactive microbial metabolites, sensitive fingerprints of microbial function, can act as inter-kingdom signaling messengers via penetration into host blood circulation and tissues. These fingerprints may be used for diagnostic purposes, and increased understanding of strain specificity in producing microbial metabolites can identify bacterial strains or specific metabolites that can be used for therapeutic purposes. Here, we will review data supporting the causal role of the gut microbiota in metabolism and discuss mechanisms and potential clinical implications.


Asunto(s)
Bacterias/metabolismo , Bacterias/patogenicidad , Microbioma Gastrointestinal , Enfermedades Metabólicas/etiología , Humanos , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología
10.
Nature ; 595(7866): 272-277, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34163067

RESUMEN

Diet is a major factor that shapes the gut microbiome1, but the consequences of diet-induced changes in the microbiome for host pathophysiology remain poorly understood. We conducted a randomized human intervention study using a very-low-calorie diet (NCT01105143). Although metabolic health was improved, severe calorie restriction led to a decrease in bacterial abundance and restructuring of the gut microbiome. Transplantation of post-diet microbiota to mice decreased their body weight and adiposity relative to mice that received pre-diet microbiota. Weight loss was associated with impaired nutrient absorption and enrichment in Clostridioides difficile, which was consistent with a decrease in bile acids and was sufficient to replicate metabolic phenotypes in mice in a toxin-dependent manner. These results emphasize the importance of diet-microbiome interactions in modulating host energy balance and the need to understand the role of diet in the interplay between pathogenic and beneficial symbionts.


Asunto(s)
Bacterias/aislamiento & purificación , Bacterias/metabolismo , Restricción Calórica , Dieta Reductora , Microbioma Gastrointestinal/fisiología , Adiposidad , Animales , Bacterias/crecimiento & desarrollo , Bacterias/patogenicidad , Toxinas Bacterianas/metabolismo , Ácidos y Sales Biliares/metabolismo , Peso Corporal , Clostridioides difficile/crecimiento & desarrollo , Clostridioides difficile/aislamiento & purificación , Clostridioides difficile/metabolismo , Metabolismo Energético , Humanos , Absorción Intestinal , Masculino , Ratones , Nutrientes/metabolismo , Simbiosis , Pérdida de Peso
11.
Physiol Rev ; 99(4): 1877-2013, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31460832

RESUMEN

The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson's disease, and Alzheimer's disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.


Asunto(s)
Bacterias/metabolismo , Encefalopatías/microbiología , Encéfalo/microbiología , Microbioma Gastrointestinal , Intestinos/microbiología , Factores de Edad , Envejecimiento , Animales , Bacterias/inmunología , Bacterias/patogenicidad , Conducta , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Encefalopatías/metabolismo , Encefalopatías/fisiopatología , Encefalopatías/psicología , Disbiosis , Sistema Nervioso Entérico/metabolismo , Sistema Nervioso Entérico/microbiología , Sistema Nervioso Entérico/fisiopatología , Interacciones Huésped-Patógeno , Humanos , Intestinos/inmunología , Neuroinmunomodulación , Plasticidad Neuronal , Factores de Riesgo
12.
Immunity ; 46(3): 421-432, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28314592

RESUMEN

How the number of immune cells recruited to sites of infection is determined and adjusted to differences in the cellular stoichiometry between host and pathogen is unknown. Here, we have uncovered a role for reactive oxygen species (ROS) as sensors of microbe size. By sensing the differential localization of ROS generated in response to microbes of different size, neutrophils tuned their interleukin (IL)-1ß expression via the selective oxidation of NF-κB, in order to implement distinct inflammatory programs. Small microbes triggered ROS intracellularly, suppressing IL-1ß expression to limit neutrophil recruitment as each phagocyte eliminated numerous pathogens. In contrast, large microbes triggered ROS extracellularly, amplifying IL-1ß expression to recruit numerous neutrophils forming cooperative clusters. Defects in ROS-mediated microbe size sensing resulted in large neutrophil infiltrates and clusters in response to small microbes that contribute to inflammatory disease. These findings highlight the impact of ROS localization on signal transduction.


Asunto(s)
Infecciones Bacterianas/inmunología , Inflamación/microbiología , Micosis/inmunología , Neutrófilos/inmunología , Especies Reactivas de Oxígeno/inmunología , Animales , Bacterias/inmunología , Bacterias/patogenicidad , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Hongos Mitospóricos/inmunología , Hongos Mitospóricos/patogenicidad , Infiltración Neutrófila/inmunología
13.
Mol Cell ; 70(5): 768-784, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29398446

RESUMEN

Bacterial toxin-antitoxin (TA) modules are abundant genetic elements that encode a toxin protein capable of inhibiting cell growth and an antitoxin that counteracts the toxin. The majority of toxins are enzymes that interfere with translation or DNA replication, but a wide variety of molecular activities and cellular targets have been described. Antitoxins are proteins or RNAs that often control their cognate toxins through direct interactions and, in conjunction with other signaling elements, through transcriptional and translational regulation of TA module expression. Three major biological functions of TA modules have been discovered, post-segregational killing ("plasmid addiction"), abortive infection (bacteriophage immunity through altruistic suicide), and persister formation (antibiotic tolerance through dormancy). In this review, we summarize the current state of the field and highlight how multiple levels of regulation shape the conditions of toxin activation to achieve the different biological functions of TA modules.


Asunto(s)
Antitoxinas/metabolismo , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , ARN Bacteriano/metabolismo , Antitoxinas/química , Antitoxinas/genética , Bacterias/genética , Bacterias/inmunología , Bacterias/patogenicidad , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Evolución Molecular , Regulación Bacteriana de la Expresión Génica , Inmunidad Innata , Viabilidad Microbiana , Modelos Moleculares , Conformación de Ácido Nucleico , Conformación Proteica , Procesamiento Postranscripcional del ARN , ARN Bacteriano/química , ARN Bacteriano/genética , Relación Estructura-Actividad , Transcripción Genética
14.
Annu Rev Microbiol ; 74: 655-671, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32689914

RESUMEN

Human-adapted bacterial pathogens use a mechanism called phase variation to randomly switch the expression of individual genes to generate a phenotypically diverse population to adapt to challenges within and between human hosts. There are increasing reports of restriction-modification systems that exhibit phase-variable expression. The outcome of phase variation of these systems is global changes in DNA methylation. Analysis of phase-variable Type I and Type III restriction-modification systems in multiple human-adapted bacterial pathogens has demonstrated that global changes in methylation regulate the expression of multiple genes. These systems are called phasevarions (phase-variable regulons). Phasevarion switching alters virulence phenotypes and facilitates evasion of host immune responses. This review describes the characteristics of phasevarions and implications for pathogenesis and immune evasion. We present and discuss examples of phasevarion systems in the major human pathogens Haemophilus influenzae, Neisseria meningitidis, Neisseria gonorrhoeae, Helicobacter pylori, Moraxella catarrhalis, and Streptococcus pneumoniae.


Asunto(s)
Bacterias/genética , Bacterias/patogenicidad , Epigénesis Genética , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Evasión Inmune , Metilación de ADN , Enzimas de Restricción-Modificación del ADN/genética , Enzimas de Restricción-Modificación del ADN/metabolismo , Humanos , Regulón , Virulencia
15.
Annu Rev Microbiol ; 74: 409-430, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32667838

RESUMEN

Bacteria produce a multitude of volatile compounds. While the biological functions of these deceptively simple molecules are unknown in many cases, for compounds that have been characterized, it is clear that they serve impressively diverse purposes. Here, we highlight recent studies that are uncovering the volatile repertoire of bacteria, and the functional relevance and impact of these molecules. We present work showing the ability of volatile compounds to modulate nutrient availability in the environment; alter the growth, development, and motility of bacteria and fungi; influence protist and arthropod behavior; and impact plant and animal health. We further discuss the benefits associated with using volatile compounds for communication and competition, alongside the challenges of studying these molecules and their functional roles. Finally, we address the opportunities these compounds present from commercial, clinical, and agricultural perspectives.


Asunto(s)
Bacterias/metabolismo , Interacciones Microbianas , Compuestos Orgánicos Volátiles/metabolismo , Bacterias/crecimiento & desarrollo , Bacterias/patogenicidad , Fenómenos Fisiológicos Bacterianos , Agentes de Control Biológico , Eucariontes/fisiología , Hongos/crecimiento & desarrollo , Hongos/metabolismo , Plantas/microbiología , Compuestos Orgánicos Volátiles/química
16.
Annu Rev Microbiol ; 74: 201-219, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32660382

RESUMEN

Bacteria are highly interactive and possess an extraordinary repertoire of intercellular communication and social behaviors, including quorum sensing (QS). QS has been studied in detail at the molecular level, so mechanistic details are well understood in many species and are often involved in virulence. The use of different animal host models has demonstrated QS-dependent control of virulence determinants and virulence in several human pathogenic bacteria. QS also controls virulence in several plant pathogenic species. Despite the role QS plays in virulence during animal and plant laboratory-engineered infections, QS mutants are frequently isolated from natural infections, demonstrating that the function of QS during infection and its role in pathogenesis remain poorly understood and are fruitful areas for future research. We discuss the role of QS during infection in various organisms and highlight approaches to better understand QS during human infection. This is an important consideration in an era of growing antimicrobial resistance, when we are looking for new ways to target bacterial infections.


Asunto(s)
Bacterias/genética , Bacterias/metabolismo , Infecciones Bacterianas/metabolismo , Percepción de Quorum/fisiología , Transducción de Señal , Animales , Bacterias/patogenicidad , Infecciones Bacterianas/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Regulación Bacteriana de la Expresión Génica , Percepción de Quorum/genética , Virulencia , Factores de Virulencia
19.
Mol Cell ; 66(6): 750-760, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28622520

RESUMEN

Infections can cause a multitude of stresses on the host and microbe. To detect potential infections, the mammalian immune system utilizes several families of pattern recognition receptors, which survey the intracellular and extracellular environments for microbial products. Members of each receptor family induce antimicrobial effector responses, which include inflammatory cytokine or interferon expression, downregulation of protein synthesis, or host cell death. In this review, we discuss the benefits of each of these innate immune responses. We highlight how non-infectious bacteria and viruses typically activate a single family of receptors, which results in a predictable host response. Infections with virulent pathogens, in contrast, may activate receptors from distinct families. As each receptor family may induce responses that antagonize or synergize with the activities of another family, cell fate decisions during pathogenic encounters are unpredictable. Understanding the antagonistic antimicrobial activities of the innate immune system should provide insight into how cell fate decisions are made during infections and potentially during other environmental stresses.


Asunto(s)
Bacterias/metabolismo , Infecciones Bacterianas/metabolismo , Inmunidad Innata , Receptores Inmunológicos/metabolismo , Transducción de Señal , Virosis/metabolismo , Virus/metabolismo , Animales , Bacterias/inmunología , Bacterias/patogenicidad , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Interacciones Huésped-Patógeno , Humanos , Ligandos , Fagocitos/inmunología , Fagocitos/metabolismo , Fagocitos/microbiología , Fagocitos/virología , Fagocitosis , Piroptosis , Receptores Inmunológicos/inmunología , Virulencia , Virosis/inmunología , Virosis/microbiología , Virus/inmunología , Virus/patogenicidad
20.
Proc Natl Acad Sci U S A ; 119(14): e2112886119, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35363569

RESUMEN

Bacterial pathogen identification, which is critical for human health, has historically relied on culturing organisms from clinical specimens. More recently, the application of machine learning (ML) to whole-genome sequences (WGSs) has facilitated pathogen identification. However, relying solely on genetic information to identify emerging or new pathogens is fundamentally constrained, especially if novel virulence factors exist. In addition, even WGSs with ML pipelines are unable to discern phenotypes associated with cryptic genetic loci linked to virulence. Here, we set out to determine if ML using phenotypic hallmarks of pathogenesis could assess potential pathogenic threat without using any sequence-based analysis. This approach successfully classified potential pathogenetic threat associated with previously machine-observed and unobserved bacteria with 99% and 85% accuracy, respectively. This work establishes a phenotype-based pipeline for potential pathogenic threat assessment, which we term PathEngine, and offers strategies for the identification of bacterial pathogens.


Asunto(s)
Bacterias , Genoma Bacteriano , Aprendizaje Automático , Factores de Virulencia , Secuenciación Completa del Genoma , Bacterias/genética , Bacterias/patogenicidad , Fenotipo , Virulencia/genética , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA