Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 115.428
Filtrar
Más filtros

Publication year range
1.
Annu Rev Immunol ; 42(1): 259-288, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38277692

RESUMEN

Gastrointestinal nematode (GIN) infection has applied significant evolutionary pressure to the mammalian immune system and remains a global economic and human health burden. Upon infection, type 2 immune sentinels activate a common antihelminth response that mobilizes and remodels the intestinal tissue for effector function; however, there is growing appreciation of the impact GIN infection also has on the distal tissue immune state. Indeed, this effect is observed even in tissues through which GINs never transit. This review highlights how GIN infection modulates systemic immunity through (a) induction of host resistance and tolerance responses, (b) secretion of immunomodulatory products, and (c) interaction with the intestinal microbiome. It also discusses the direct consequences that changes to distal tissue immunity can have for concurrent and subsequent infection, chronic noncommunicable diseases, and vaccination efficacy.


Asunto(s)
Microbioma Gastrointestinal , Nematodos , Infecciones por Nematodos , Animales , Humanos , Infecciones por Nematodos/inmunología , Nematodos/inmunología , Nematodos/fisiología , Microbioma Gastrointestinal/inmunología , Inmunomodulación , Interacciones Huésped-Parásitos/inmunología , Parasitosis Intestinales/inmunología , Tolerancia Inmunológica , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/parasitología
2.
Annu Rev Immunol ; 41: 343-373, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-36750314

RESUMEN

A large body of evidence generated in the last two and a half years addresses the roles of T cells in SARS-CoV-2 infection and following vaccination. Infection or vaccination induces multi-epitope CD4 and CD8 T cell responses with polyfunctionality. Early T cell responses have been associated with mild COVID-19 outcomes. In concert with animal model data, these results suggest that while antibody responses are key to prevent infection, T cell responses may also play valuable roles in reducing disease severity and controlling infection. T cell memory after vaccination is sustained for at least six months. While neutralizing antibody responses are impacted by SARS-CoV-2 variants, most CD4 and CD8 T cell responses are preserved. This review highlights the extensive progress made, and the data and knowledge gaps that remain, in our understanding of T cell responses to SARS-CoV-2 and COVID-19 vaccines.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Humanos , Vacunas contra la COVID-19 , Linfocitos T CD8-positivos , Anticuerpos Antivirales
3.
Annu Rev Immunol ; 40: 499-523, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35471839

RESUMEN

The bladder is a major component of the urinary tract, an organ system that expels metabolic waste and excess water, which necessitates proximity to the external environment and its pathogens. It also houses a commensal microbiome. Therefore, its tissue immunity must resist pathogen invasion while maintaining tolerance to commensals. Bacterial infection of the bladder is common, with half of women globally experiencing one or more episodes of cystitis in their lifetime. Despite this, our knowledge of bladder immunity, particularly in humans, is incomplete. Here we consider the current view of tissue immunity in the bladder, with a focus on defense against infection. The urothelium has robust immune functionality, and its defensive capabilities are supported by resident immune cells, including macrophages, dendritic cells, natural killer cells, and γδ T cells. We discuss each in turn and consider why adaptive immune responses are often ineffective in preventing recurrent infection, as well as areas of priority for future research.


Asunto(s)
Infecciones Bacterianas , Vejiga Urinaria , Animales , Femenino , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Macrófagos , Vejiga Urinaria/microbiología
4.
Annu Rev Immunol ; 38: 203-228, 2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31986071

RESUMEN

Mucosal-associated invariant T (MAIT) cells have been attracting increasing attention over the last few years as a potent unconventional T cell subset. Three factors largely account for this emerging interest. Firstly, these cells are abundant in humans, both in circulation and especially in some tissues such as the liver. Secondly is the discovery of a ligand that has uncovered their microbial targets, and also allowed for the development of tools to accurately track the cells in both humans and mice. Finally, it appears that the cells not only have a diverse range of functions but also are sensitive to a range of inflammatory triggers that can enhance or even bypass T cell receptor-mediated signals-substantially broadening their likely impact in health and disease. In this review we discuss how MAIT cells display antimicrobial, homeostatic, and amplifier roles in vivo, and how this may lead to protection and potentially pathology.


Asunto(s)
Susceptibilidad a Enfermedades , Homeostasis , Células T Invariantes Asociadas a Mucosa/inmunología , Células T Invariantes Asociadas a Mucosa/metabolismo , Animales , Biomarcadores , Interacciones Huésped-Patógeno , Humanos , Inmunidad Mucosa , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Membrana Mucosa/microbiología , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
5.
Annu Rev Immunol ; 38: 79-98, 2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31800327

RESUMEN

DNA has been known to be a potent immune stimulus for more than half a century. However, the underlying molecular mechanisms of DNA-triggered immune response have remained elusive until recent years. Cyclic GMP-AMP synthase (cGAS) is a major cytoplasmic DNA sensor in various types of cells that detect either invaded foreign DNA or aberrantly located self-DNA. Upon sensing of DNA, cGAS catalyzes the formation of cyclic GMP-AMP (cGAMP), which in turn activates the ER-localized adaptor protein MITA (also named STING) to elicit the innate immune response. The cGAS-MITA axis not only plays a central role in host defense against pathogen-derived DNA but also acts as a cellular stress response pathway by sensing aberrantly located self-DNA, which is linked to the pathogenesis of various human diseases. In this review, we summarize the spatial and temporal mechanisms of host defense to cytoplasmic DNA mediated by the cGAS-MITA axis and discuss the association of malfunctions of this axis with autoimmune and other diseases.


Asunto(s)
ADN/inmunología , Inmunidad Innata , Animales , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Autoinmunidad , Biomarcadores , Citoplasma/inmunología , Citoplasma/metabolismo , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno/inmunología , Humanos , Evasión Inmune , Interferón Tipo I/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo
6.
Annu Rev Immunol ; 37: 547-570, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30699000

RESUMEN

Adaptive immune recognition is mediated by antigen receptors on B and T cells generated by somatic recombination during lineage development. The high level of diversity resulting from this process posed technical limitations that previously limited the comprehensive analysis of adaptive immune recognition. Advances over the last ten years have produced data and approaches allowing insights into how T cells develop, evolutionary signatures of recombination and selection, and the features of T cell receptors that mediate epitope-specific binding and T cell activation. The size and complexity of these data have necessitated the generation of novel computational and analytical approaches, which are transforming how T cell immunology is conducted. Here we review the development and application of novel biological, theoretical, and computational methods for understanding T cell recognition and discuss the potential for improved models of receptor:antigen interactions.


Asunto(s)
Biología Computacional/métodos , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Antígenos/inmunología , Antígenos/metabolismo , Diferenciación Celular , Selección Clonal Mediada por Antígenos , Epítopos de Linfocito T/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/metabolismo
7.
Annu Rev Immunol ; 37: 405-437, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30673535

RESUMEN

Pathogenic organisms exert a negative impact on host health, revealed by the clinical signs of infectious diseases. Immunity limits the severity of infectious diseases through resistance mechanisms that sense and target pathogens for containment, killing, or expulsion. These resistance mechanisms are viewed as the prevailing function of immunity. Under pathophysiologic conditions, however, immunity arises in response to infections that carry health and fitness costs to the host. Therefore, additional defense mechanisms are required to limit these costs, before immunity becomes operational as well as thereafter to avoid immunopathology. These are tissue damage control mechanisms that adjust the metabolic output of host tissues to different forms of stress and damage associated with infection. Disease tolerance is the term used to define this defense strategy, which does not exert a direct impact on pathogens but is essential to limit the health and fitness costs of infection. Under this argument, we propose that disease tolerance is an inherent component of immunity.


Asunto(s)
Resistencia a la Enfermedad/inmunología , Inmunidad Innata , Infecciones/inmunología , Microbiota/inmunología , Animales , Interacciones Huésped-Patógeno , Humanos , Tolerancia Inmunológica , Inmunomodulación
8.
Annu Rev Immunol ; 36: 603-638, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29490165

RESUMEN

Globally, about 36.7 million people were living with HIV infection at the end of 2015. The most frequent infection co-occurring with HIV-1 is Mycobacterium tuberculosis-374,000 deaths per annum are attributable to HIV-tuberculosis, 75% of those occurring in Africa. HIV-1 infection increases the risk of tuberculosis by a factor of up to 26 and alters its clinical presentation, complicates diagnosis and treatment, and worsens outcome. Although HIV-1-induced depletion of CD4+ T cells underlies all these effects, more widespread immune deficits also contribute to susceptibility and pathogenesis. These defects present a challenge to understand and ameliorate, but also an opportunity to learn and optimize mechanisms that normally protect people against tuberculosis. The most effective means to prevent and ameliorate tuberculosis in HIV-1-infected people is antiretroviral therapy, but this may be complicated by pathological immune deterioration that in turn requires more effective host-directed anti-inflammatory therapies to be derived.


Asunto(s)
Coinfección , Infecciones por VIH/inmunología , VIH-1/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Animales , Terapia Antirretroviral Altamente Activa , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Progresión de la Enfermedad , Variación Genética , Infecciones por VIH/diagnóstico , Infecciones por VIH/terapia , Infecciones por VIH/virología , VIH-1/genética , Humanos , Tuberculosis/diagnóstico , Tuberculosis/microbiología , Tuberculosis/terapia , Replicación Viral
9.
Annu Rev Immunol ; 36: 309-338, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29677470

RESUMEN

The complement system is an evolutionarily ancient key component of innate immunity required for the detection and removal of invading pathogens. It was discovered more than 100 years ago and was originally defined as a liver-derived, blood-circulating sentinel system that classically mediates the opsonization and lytic killing of dangerous microbes and the initiation of the general inflammatory reaction. More recently, complement has also emerged as a critical player in adaptive immunity via its ability to instruct both B and T cell responses. In particular, work on the impact of complement on T cell responses led to the surprising discoveries that the complement system also functions within cells and is involved in regulating basic cellular processes, predominantly those of metabolic nature. Here, we review current knowledge about complement's role in T cell biology, with a focus on the novel intracellular and noncanonical activities of this ancient system.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Inmunomodulación , Linfocitos T/inmunología , Linfocitos T/metabolismo , Inmunidad Adaptativa , Animales , Autoinmunidad , Linfocitos B/inmunología , Linfocitos B/metabolismo , Activación de Complemento/inmunología , Metabolismo Energético , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Celular , Proteína Cofactora de Membrana/metabolismo , Células TH1/inmunología , Células TH1/metabolismo
10.
Cell ; 187(9): 2079-2094, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38670066

RESUMEN

Several conceptual pillars form the foundation of modern immunology, including the clonal selection theory, antigen receptor diversity, immune memory, and innate control of adaptive immunity. However, some immunological phenomena cannot be explained by the current framework. Thus, we still do not know how to design vaccines that would provide long-lasting protective immunity against certain pathogens, why autoimmune responses target some antigens and not others, or why the immune response to infection sometimes does more harm than good. Understanding some of these mysteries may require that we question existing assumptions to develop and test alternative explanations. Immunology is increasingly at a point when, once again, exploring new perspectives becomes a necessity.


Asunto(s)
Alergia e Inmunología , Humanos , Animales , Alergia e Inmunología/tendencias , Inmunidad Adaptativa , Inmunidad Innata , Memoria Inmunológica
11.
Cell ; 187(2): 390-408.e23, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-38157855

RESUMEN

We describe a human lung disease caused by autosomal recessive, complete deficiency of the monocyte chemokine receptor C-C motif chemokine receptor 2 (CCR2). Nine children from five independent kindreds have pulmonary alveolar proteinosis (PAP), progressive polycystic lung disease, and recurrent infections, including bacillus Calmette Guérin (BCG) disease. The CCR2 variants are homozygous in six patients and compound heterozygous in three, and all are loss-of-expression and loss-of-function. They abolish CCR2-agonist chemokine C-C motif ligand 2 (CCL-2)-stimulated Ca2+ signaling in and migration of monocytic cells. All patients have high blood CCL-2 levels, providing a diagnostic test for screening children with unexplained lung or mycobacterial disease. Blood myeloid and lymphoid subsets and interferon (IFN)-γ- and granulocyte-macrophage colony-stimulating factor (GM-CSF)-mediated immunity are unaffected. CCR2-deficient monocytes and alveolar macrophage-like cells have normal gene expression profiles and functions. By contrast, alveolar macrophage counts are about half. Human complete CCR2 deficiency is a genetic etiology of PAP, polycystic lung disease, and recurrent infections caused by impaired CCL2-dependent monocyte migration to the lungs and infected tissues.


Asunto(s)
Proteinosis Alveolar Pulmonar , Receptores CCR2 , Niño , Humanos , Pulmón/metabolismo , Macrófagos Alveolares/metabolismo , Proteinosis Alveolar Pulmonar/genética , Proteinosis Alveolar Pulmonar/diagnóstico , Receptores CCR2/deficiencia , Receptores CCR2/genética , Receptores CCR2/metabolismo , Reinfección/metabolismo
12.
Cell ; 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38897196

RESUMEN

Reversing CD8+ T cell dysfunction is crucial in treating chronic hepatitis B virus (HBV) infection, yet specific molecular targets remain unclear. Our study analyzed co-signaling receptors during hepatocellular priming and traced the trajectory and fate of dysfunctional HBV-specific CD8+ T cells. Early on, these cells upregulate PD-1, CTLA-4, LAG-3, OX40, 4-1BB, and ICOS. While blocking co-inhibitory receptors had minimal effect, activating 4-1BB and OX40 converted them into antiviral effectors. Prolonged stimulation led to a self-renewing, long-lived, heterogeneous population with a unique transcriptional profile. This includes dysfunctional progenitor/stem-like (TSL) cells and two distinct dysfunctional tissue-resident memory (TRM) populations. While 4-1BB expression is ubiquitously maintained, OX40 expression is limited to TSL. In chronic settings, only 4-1BB stimulation conferred antiviral activity. In HBeAg+ chronic patients, 4-1BB activation showed the highest potential to rejuvenate dysfunctional CD8+ T cells. Targeting all dysfunctional T cells, rather than only stem-like precursors, holds promise for treating chronic HBV infection.

13.
Cell ; 187(8): 1874-1888.e14, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38518773

RESUMEN

Infections of the lung cause observable sickness thought to be secondary to inflammation. Signs of sickness are crucial to alert others via behavioral-immune responses to limit contact with contagious individuals. Gram-negative bacteria produce exopolysaccharide (EPS) that provides microbial protection; however, the impact of EPS on sickness remains uncertain. Using genome-engineered Pseudomonas aeruginosa (P. aeruginosa) strains, we compared EPS-producers versus non-producers and a virulent Escherichia coli (E. coli) lung infection model in male and female mice. EPS-negative P. aeruginosa and virulent E. coli infection caused severe sickness, behavioral alterations, inflammation, and hypothermia mediated by TLR4 detection of the exposed lipopolysaccharide (LPS) in lung TRPV1+ sensory neurons. However, inflammation did not account for sickness. Stimulation of lung nociceptors induced acute stress responses in the paraventricular hypothalamic nuclei by activating corticotropin-releasing hormone neurons responsible for sickness behavior and hypothermia. Thus, EPS-producing biofilm pathogens evade initiating a lung-brain sensory neuronal response that results in sickness.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli , Pulmón , Polisacáridos Bacterianos , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Animales , Femenino , Masculino , Ratones , Biopelículas , Escherichia coli/fisiología , Hipotermia/metabolismo , Hipotermia/patología , Inflamación/metabolismo , Inflamación/patología , Pulmón/microbiología , Pulmón/patología , Neumonía/microbiología , Neumonía/patología , Pseudomonas aeruginosa/fisiología , Células Receptoras Sensoriales , Polisacáridos Bacterianos/metabolismo , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Nociceptores/metabolismo
14.
Cell ; 186(12): 2690-2704.e20, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37295405

RESUMEN

Biofilm formation is generally recognized as a bacterial defense mechanism against environmental threats, including antibiotics, bacteriophages, and leukocytes of the human immune system. Here, we show that for the human pathogen Vibrio cholerae, biofilm formation is not only a protective trait but also an aggressive trait to collectively predate different immune cells. We find that V. cholerae forms biofilms on the eukaryotic cell surface using an extracellular matrix comprising primarily mannose-sensitive hemagglutinin pili, toxin-coregulated pili, and the secreted colonization factor TcpF, which differs from the matrix composition of biofilms on other surfaces. These biofilms encase immune cells and establish a high local concentration of a secreted hemolysin to kill the immune cells before the biofilms disperse in a c-di-GMP-dependent manner. Together, these results uncover how bacteria employ biofilm formation as a multicellular strategy to invert the typical relationship between human immune cells as the hunters and bacteria as the hunted.


Asunto(s)
Vibrio cholerae , Animales , Humanos , Vibrio cholerae/metabolismo , Conducta Predatoria , Biopelículas , Fimbrias Bacterianas , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica
15.
Cell ; 186(25): 5486-5499.e13, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-37951212

RESUMEN

Germinal centers (GCs) form in lymph nodes after immunization or infection to facilitate antibody affinity maturation and memory and plasma cell (PC) development. PC differentiation is thought to involve stringent selection for GC B cells expressing the highest-affinity antigen receptors, but how this plays out during complex polyclonal responses is unclear. We combine temporal lineage tracing with antibody characterization to gain a snapshot of PCs developing during influenza infection. GCs co-mature B cell clones with antibody affinities spanning multiple orders of magnitude; however, each generates PCs with similar efficiencies, including weak binders. Within lineages, PC selection is not restricted to variants with the highest-affinity antibodies. Differentiation is commonly associated with proliferative expansion to produce "nodes" of identical PCs. Immunization-induced GCs generate fewer PCs but still of low- and high-antibody affinities. We propose that generating low-affinity antibody PCs reflects an evolutionary compromise to facilitate diverse serum antibody responses.


Asunto(s)
Afinidad de Anticuerpos , Linfocitos B , Centro Germinal , Células Plasmáticas , Formación de Anticuerpos , Linfocitos B/citología , Linfocitos B/inmunología , Ganglios Linfáticos , Línea Celular , Humanos , Animales , Ratones , Cricetinae , Virus de la Influenza A/inmunología , Diferenciación Celular
16.
Cell ; 186(21): 4632-4651.e23, 2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37776858

RESUMEN

The dynamics of immunity to infection in infants remain obscure. Here, we used a multi-omics approach to perform a longitudinal analysis of immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in infants and young children by analyzing blood samples and weekly nasal swabs collected before, during, and after infection with Omicron and non-Omicron variants. Infection stimulated robust antibody titers that, unlike in adults, showed no sign of decay for up to 300 days. Infants mounted a robust mucosal immune response characterized by inflammatory cytokines, interferon (IFN) α, and T helper (Th) 17 and neutrophil markers (interleukin [IL]-17, IL-8, and CXCL1). The immune response in blood was characterized by upregulation of activation markers on innate cells, no inflammatory cytokines, but several chemokines and IFNα. The latter correlated with viral load and expression of interferon-stimulated genes (ISGs) in myeloid cells measured by single-cell multi-omics. Together, these data provide a snapshot of immunity to infection during the initial weeks and months of life.


Asunto(s)
COVID-19 , SARS-CoV-2 , Adulto , Niño , Lactante , Humanos , Preescolar , SARS-CoV-2/metabolismo , Multiómica , Citocinas/metabolismo , Interferón-alfa , Inmunidad Mucosa
17.
Cell ; 186(19): 4059-4073.e27, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37611581

RESUMEN

Antimicrobial resistance is a leading mortality factor worldwide. Here, we report the discovery of clovibactin, an antibiotic isolated from uncultured soil bacteria. Clovibactin efficiently kills drug-resistant Gram-positive bacterial pathogens without detectable resistance. Using biochemical assays, solid-state nuclear magnetic resonance, and atomic force microscopy, we dissect its mode of action. Clovibactin blocks cell wall synthesis by targeting pyrophosphate of multiple essential peptidoglycan precursors (C55PP, lipid II, and lipid IIIWTA). Clovibactin uses an unusual hydrophobic interface to tightly wrap around pyrophosphate but bypasses the variable structural elements of precursors, accounting for the lack of resistance. Selective and efficient target binding is achieved by the sequestration of precursors into supramolecular fibrils that only form on bacterial membranes that contain lipid-anchored pyrophosphate groups. This potent antibiotic holds the promise of enabling the design of improved therapeutics that kill bacterial pathogens without resistance development.


Asunto(s)
Antibacterianos , Bacterias , Microbiología del Suelo , Antibacterianos/aislamiento & purificación , Antibacterianos/farmacología , Bioensayo , Difosfatos
18.
Cell ; 186(9): 1877-1894.e27, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37116470

RESUMEN

Negative-stranded RNA viruses can establish long-term persistent infection in the form of large intracellular inclusions in the human host and cause chronic diseases. Here, we uncover how cellular stress disrupts the metastable host-virus equilibrium in persistent infection and induces viral replication in a culture model of mumps virus. Using a combination of cell biology, whole-cell proteomics, and cryo-electron tomography, we show that persistent viral replication factories are dynamic condensates and identify the largely disordered viral phosphoprotein as a driver of their assembly. Upon stress, increased phosphorylation of the phosphoprotein at its interaction interface with the viral polymerase coincides with the formation of a stable replication complex. By obtaining atomic models for the authentic mumps virus nucleocapsid, we elucidate a concomitant conformational change that exposes the viral genome to its replication machinery. These events constitute a stress-mediated switch within viral condensates that provide an environment to support upregulation of viral replication.


Asunto(s)
Virus de la Parotiditis , Infección Persistente , Humanos , Virus de la Parotiditis/fisiología , Nucleocápside , Fosfoproteínas/metabolismo , Replicación Viral
19.
Cell ; 186(9): 1863-1876.e16, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37030292

RESUMEN

Over the past few years, numerous anti-phage defense systems have been discovered in bacteria. Although the mechanism of defense for some of these systems is understood, a major unanswered question is how these systems sense phage infection. To systematically address this question, we isolated 177 phage mutants that escape 15 different defense systems. In many cases, these escaper phages were mutated in the gene sensed by the defense system, enabling us to map the phage determinants that confer sensitivity to bacterial immunity. Our data identify specificity determinants of diverse retron systems and reveal phage-encoded triggers for multiple abortive infection systems. We find general themes in phage sensing and demonstrate that mechanistically diverse systems have converged to sense either the core replication machinery of the phage, phage structural components, or host takeover mechanisms. Combining our data with previous findings, we formulate key principles on how bacterial immune systems sense phage invaders.


Asunto(s)
Bacterias , Bacteriófagos , Bacterias/genética , Bacterias/virología , Bacteriófagos/genética , Sistemas CRISPR-Cas , Proteínas Virales/metabolismo , Mutación , Fenómenos Fisiológicos Bacterianos
20.
Cell ; 186(18): 3882-3902.e24, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37597510

RESUMEN

Inflammation can trigger lasting phenotypes in immune and non-immune cells. Whether and how human infections and associated inflammation can form innate immune memory in hematopoietic stem and progenitor cells (HSPC) has remained unclear. We found that circulating HSPC, enriched from peripheral blood, captured the diversity of bone marrow HSPC, enabling investigation of their epigenomic reprogramming following coronavirus disease 2019 (COVID-19). Alterations in innate immune phenotypes and epigenetic programs of HSPC persisted for months to 1 year following severe COVID-19 and were associated with distinct transcription factor (TF) activities, altered regulation of inflammatory programs, and durable increases in myelopoiesis. HSPC epigenomic alterations were conveyed, through differentiation, to progeny innate immune cells. Early activity of IL-6 contributed to these persistent phenotypes in human COVID-19 and a mouse coronavirus infection model. Epigenetic reprogramming of HSPC may underlie altered immune function following infection and be broadly relevant, especially for millions of COVID-19 survivors.


Asunto(s)
COVID-19 , Memoria Epigenética , Síndrome Post Agudo de COVID-19 , Animales , Humanos , Ratones , Diferenciación Celular , COVID-19/inmunología , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas , Inflamación/genética , Inmunidad Entrenada , Monocitos/inmunología , Síndrome Post Agudo de COVID-19/genética , Síndrome Post Agudo de COVID-19/inmunología , Síndrome Post Agudo de COVID-19/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda