Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.398
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 40: 271-294, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35080919

RESUMEN

Vertebrate immune systems suppress viral infection using both innate restriction factors and adaptive immunity. Viruses mutate to escape these defenses, driving hosts to counterevolve to regain fitness. This cycle recurs repeatedly, resulting in an evolutionary arms race whose outcome depends on the pace and likelihood of adaptation by host and viral genes. Although viruses evolve faster than their vertebrate hosts, their proteins are subject to numerous functional constraints that impact the probability of adaptation. These constraints are globally defined by evolutionary landscapes, which describe the fitness and adaptive potential of all possible mutations. We review deep mutational scanning experiments mapping the evolutionary landscapes of both host and viral proteins engaged in arms races. For restriction factors and some broadly neutralizing antibodies, landscapes favor the host, which may help to level the evolutionary playing field against rapidly evolving viruses. We discuss the biophysical underpinnings of these landscapes and their therapeutic implications.


Asunto(s)
Virosis , Virus , Animales , Evolución Biológica , Humanos , Mutación , Proteínas Virales , Virosis/genética , Virus/genética
2.
Annu Rev Immunol ; 37: 247-267, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30633609

RESUMEN

Recognition of foreign nucleic acids is the primary mechanism by which a type I interferon-mediated antiviral response is triggered. Given that human cells are replete with DNA and RNA, this evolutionary strategy poses an inherent biological challenge, i.e., the fundamental requirement to reliably differentiate self-nucleic acids from nonself nucleic acids. We suggest that the group of Mendelian inborn errors of immunity referred to as the type I interferonopathies relate to a breakdown of self/nonself discrimination, with the associated mutant genotypes involving molecules playing direct or indirect roles in nucleic acid signaling. This perspective begs the question as to the sources of self-derived nucleic acids that drive an inappropriate immune response. Resolving this question will provide fundamental insights into immune tolerance, antiviral signaling, and complex autoinflammatory disease states. Here we develop these ideas, discussing type I interferonopathies within the broader framework of nucleic acid-driven inflammation.


Asunto(s)
Antígenos Virales/inmunología , Autoantígenos/inmunología , Enfermedades del Sistema Inmune/inmunología , Ácidos Nucleicos/inmunología , Virosis/inmunología , Animales , Humanos , Enfermedades del Sistema Inmune/genética , Tolerancia Inmunológica , Inmunidad Innata , Interferón Tipo I/metabolismo , Virosis/genética
3.
Annu Rev Immunol ; 33: 79-106, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25493335

RESUMEN

Cell proliferation and cell death are integral elements in maintaining homeostatic balance in metazoans. Disease pathologies ensue when these processes are disturbed. A plethora of evidence indicates that malfunction of cell death can lead to inflammation, autoimmunity, or immunodeficiency. Programmed necrosis or necroptosis is a form of nonapoptotic cell death driven by the receptor interacting protein kinase 3 (RIPK3) and its substrate, mixed lineage kinase domain-like (MLKL). RIPK3 partners with its upstream adaptors RIPK1, TRIF, or DAI to signal for necroptosis in response to death receptor or Toll-like receptor stimulation, pathogen infection, or sterile cell injury. Necroptosis promotes inflammation through leakage of cellular contents from damaged plasma membranes. Intriguingly, many of the signal adaptors of necroptosis have dual functions in innate immune signaling. This unique signature illustrates the cooperative nature of necroptosis and innate inflammatory signaling pathways in managing cell and organismal stresses from pathogen infection and sterile tissue injury.


Asunto(s)
Inflamación/metabolismo , Inflamación/patología , Necrosis/metabolismo , Transducción de Señal , Animales , Infecciones Bacterianas/genética , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/patología , Evolución Biológica , Muerte Celular , Humanos , Inflamasomas/metabolismo , Inflamación/genética , Interleucina-1beta/metabolismo , FN-kappa B/metabolismo , Enfermedades Parasitarias/genética , Enfermedades Parasitarias/metabolismo , Enfermedades Parasitarias/patología , Fosforilación , Unión Proteica , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Ubiquitinación , Virosis/genética , Virosis/metabolismo , Virosis/patología
4.
Annu Rev Immunol ; 32: 189-225, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24423116

RESUMEN

Adoptive immunotherapy, or the infusion of lymphocytes, is a promising approach for the treatment of cancer and certain chronic viral infections. The application of the principles of synthetic biology to enhance T cell function has resulted in substantial increases in clinical efficacy. The primary challenge to the field is to identify tumor-specific targets to avoid off-tumor, on-target toxicity. Given recent advances in efficacy in numerous pilot trials, the next steps in clinical development will require multicenter trials to establish adoptive immunotherapy as a mainstream technology.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias/inmunología , Neoplasias/terapia , Virosis/inmunología , Virosis/terapia , Traslado Adoptivo , Animales , Antígenos/genética , Antígenos/inmunología , Biomarcadores , Tratamiento Basado en Trasplante de Células y Tejidos , Técnicas de Transferencia de Gen , Terapia Genética , Humanos , Neoplasias/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Transducción Genética , Virosis/genética
5.
Cell ; 181(5): 1046-1061.e6, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32392465

RESUMEN

Since their discovery, giant viruses have expanded our understanding of the principles of virology. Due to their gargantuan size and complexity, little is known about the life cycles of these viruses. To answer outstanding questions regarding giant virus infection mechanisms, we set out to determine biomolecular conditions that promote giant virus genome release. We generated four infection intermediates in Samba virus (Mimivirus genus, lineage A) as visualized by cryoelectron microscopy (cryo-EM), cryoelectron tomography (cryo-ET), and scanning electron microscopy (SEM). Each of these four intermediates reflects similar morphology to a stage that occurs in vivo. We show that these genome release stages are conserved in other mimiviruses. Finally, we identified proteins that are released from Samba and newly discovered Tupanvirus through differential mass spectrometry. Our work revealed the molecular forces that trigger infection are conserved among disparate giant viruses. This study is also the first to identify specific proteins released during the initial stages of giant virus infection.


Asunto(s)
Virus Gigantes/genética , Virus Gigantes/metabolismo , Virus Gigantes/fisiología , Cápside/metabolismo , Virus ADN/genética , Genoma Viral/genética , Proteómica/métodos , Ensamble de Virus/genética , Ensamble de Virus/fisiología , Virosis/genética , Virus/genética
6.
Cell ; 183(5): 1312-1324.e10, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33212011

RESUMEN

Interferon (IFN)-Is are crucial mediators of antiviral immunity and homeostatic immune system regulation. However, the source of IFN-I signaling under homeostatic conditions is unclear. We discovered that commensal microbes regulate the IFN-I response through induction of IFN-ß by colonic DCs. Moreover, the mechanism by which a specific commensal microbe induces IFN-ß was identified. Outer membrane (OM)-associated glycolipids of gut commensal microbes belonging to the Bacteroidetes phylum induce expression of IFN-ß. Using Bacteroides fragilis and its OM-associated polysaccharide A, we determined that IFN-ß expression was induced via TLR4-TRIF signaling. Antiviral activity of this purified microbial molecule against infection with either vesicular stomatitis virus (VSV) or influenza was demonstrated to be dependent on the induction of IFN-ß. In a murine VSV infection model, commensal-induced IFN-ß regulated natural resistance to virus infection. Due to the physiological importance of IFN-Is, discovery of an IFN-ß-inducing microbial molecule represents a potential approach for the treatment of some human diseases.


Asunto(s)
Inmunidad Innata , Microbiota , Virosis/microbiología , Animales , Bacteroides fragilis/fisiología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Colon/patología , Colon/virología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glucolípidos/metabolismo , Inmunidad Innata/efectos de los fármacos , Interferón beta/sangre , Interferón beta/metabolismo , Masculino , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Polisacáridos Bacterianos/farmacología , Receptor Toll-Like 4/metabolismo , Vesiculovirus/fisiología , Virosis/genética
7.
Nat Rev Mol Cell Biol ; 23(4): 286-301, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34815573

RESUMEN

Double-stranded RNA (dsRNA) is associated with most viral infections - it either constitutes the viral genome (in the case of dsRNA viruses) or is generated in host cells during viral replication. Hence, nearly all organisms have the capability of recognizing dsRNA and mounting a response, the primary aim of which is to mitigate the potential infection. In vertebrates, a set of innate immune receptors for dsRNA induce a multitude of cell-intrinsic and cell-extrinsic immune responses upon dsRNA recognition. Notably, recent studies showed that vertebrate cells can accumulate self-derived dsRNAs or dsRNA-like species upon dysregulation of several cellular processes, activating the very same immune pathways as in infected cells. On the one hand, such aberrant immune activation in the absence of infection can lead to pathogenesis of immune disorders, such as Aicardi-Goutières syndrome. On the other hand, the same innate immune reaction can be induced in a controlled setting for a therapeutic benefit, as occurs in immunotherapies. In this Review, we describe mechanisms by which immunostimulatory dsRNAs are generated in mammalian cells, either by viruses or by the host cells, and how cells respond to them, with the focus on recent developments regarding the role of cellular dsRNAs in immune modulation.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso , Malformaciones del Sistema Nervioso , Virosis , Animales , Inmunidad Innata , Mamíferos , ARN Bicatenario , Virosis/genética , Replicación Viral
8.
Annu Rev Biochem ; 85: 431-54, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26844395

RESUMEN

Recent developments indicate that macrodomains, an ancient and diverse protein domain family, are key players in the recognition, interpretation, and turnover of ADP-ribose (ADPr) signaling. Crucial to this is the ability of macrodomains to recognize ADPr either directly, in the form of a metabolic derivative, or as a modification covalently bound to proteins. Thus, macrodomains regulate a wide variety of cellular and organismal processes, including DNA damage repair, signal transduction, and immune response. Their importance is further indicated by the fact that dysregulation or mutation of a macrodomain is associated with several diseases, including cancer, developmental defects, and neurodegeneration. In this review, we summarize the current insights into macrodomain evolution and how this evolution influenced their structural and functional diversification. We highlight some aspects of macrodomain roles in pathobiology as well as their emerging potential as therapeutic targets.


Asunto(s)
Reparación del ADN , Proteínas de Escherichia coli/química , Neoplasias/enzimología , Poli(ADP-Ribosa) Polimerasas/química , Procesamiento Proteico-Postraduccional , Proteínas Represoras/química , Virosis/enzimología , Adenosina Difosfato Ribosa/química , Adenosina Difosfato Ribosa/metabolismo , Animales , Daño del ADN , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Evolución Molecular , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Familia de Multigenes , Neoplasias/química , Neoplasias/genética , Neoplasias/patología , Filogenia , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Dominios Proteicos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Homología Estructural de Proteína , Virosis/genética , Virosis/patología , Virosis/virología
9.
Nat Immunol ; 20(5): 637-651, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30962590

RESUMEN

Respiratory infections are common precursors to asthma exacerbations in children, but molecular immune responses that determine whether and how an infection causes an exacerbation are poorly understood. By using systems-scale network analysis, we identify repertoires of cellular transcriptional pathways that lead to and underlie distinct patterns of asthma exacerbation. Specifically, in both virus-associated and nonviral exacerbations, we demonstrate a set of core exacerbation modules, among which epithelial-associated SMAD3 signaling is upregulated and lymphocyte response pathways are downregulated early in exacerbation, followed by later upregulation of effector pathways including epidermal growth factor receptor signaling, extracellular matrix production, mucus hypersecretion, and eosinophil activation. We show an additional set of multiple inflammatory cell pathways involved in virus-associated exacerbations, in contrast to squamous cell pathways associated with nonviral exacerbations. Our work introduces an in vivo molecular platform to investigate, in a clinical setting, both the mechanisms of disease pathogenesis and therapeutic targets to modify exacerbations.


Asunto(s)
Asma/inmunología , Redes Reguladoras de Genes/inmunología , Transcriptoma/inmunología , Virosis/inmunología , Adolescente , Asma/genética , Asma/virología , Estudios de Casos y Controles , Niño , Resfriado Común/genética , Resfriado Común/inmunología , Resfriado Común/virología , Femenino , Humanos , Estudios Longitudinales , Masculino , Estudios Prospectivos , Transducción de Señal/genética , Transducción de Señal/inmunología , Virosis/genética , Virosis/virología
10.
Nat Immunol ; 20(7): 812-823, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31036902

RESUMEN

The helicase RIG-I initiates an antiviral immune response after recognition of pathogenic RNA. TRIM25, an E3 ubiquitin ligase, mediates K63-linked ubiquitination of RIG-I, which is crucial for RIG-I downstream signaling and the antiviral innate immune response. The components and mode of the RIG-I-initiated innate signaling remain to be fully understood. Here we identify a novel long noncoding RNA (Lnczc3h7a) that binds to TRIM25 and promotes RIG-I-mediated antiviral innate immune responses. Depletion of Lnczc3h7a impairs RIG-I signaling and the antiviral innate response to RNA viruses in vitro and in vivo. Mechanistically, Lnczc3h7a binds to both TRIM25 and activated RIG-I, serving as a molecular scaffold for stabilization of the RIG-I-TRIM25 complex at the early stage of viral infection. Lnczc3h7a facilitates TRIM25-mediated K63-linked ubiquitination of RIG-I and thus promotes downstream signaling transduction. Our findings reveal that host RNAs can enhance the response of innate immune sensors to foreign RNAs, ensuring effective antiviral defense.


Asunto(s)
Proteína 58 DEAD Box/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/genética , ARN Largo no Codificante/genética , Factores de Transcripción/genética , Animales , Línea Celular , Humanos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/virología , Ratones , Modelos Biológicos , Interferencia de ARN , Virus ARN/inmunología , Transducción de Señal , Virosis/genética , Virosis/inmunología , Virosis/metabolismo , Virosis/virología
11.
Cell ; 167(3): 643-656.e17, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27768888

RESUMEN

Humans differ in the outcome that follows exposure to life-threatening pathogens, yet the extent of population differences in immune responses and their genetic and evolutionary determinants remain undefined. Here, we characterized, using RNA sequencing, the transcriptional response of primary monocytes from Africans and Europeans to bacterial and viral stimuli-ligands activating Toll-like receptor pathways (TLR1/2, TLR4, and TLR7/8) and influenza virus-and mapped expression quantitative trait loci (eQTLs). We identify numerous cis-eQTLs that contribute to the marked differences in immune responses detected within and between populations and a strong trans-eQTL hotspot at TLR1 that decreases expression of pro-inflammatory genes in Europeans only. We find that immune-responsive regulatory variants are enriched in population-specific signals of natural selection and show that admixture with Neandertals introduced regulatory variants into European genomes, affecting preferentially responses to viral challenges. Together, our study uncovers evolutionarily important determinants of differences in host immune responsiveness between human populations.


Asunto(s)
Adaptación Fisiológica/genética , Adaptación Fisiológica/inmunología , Inmunidad Adaptativa , Hombre de Neandertal/genética , Hombre de Neandertal/inmunología , Inmunidad Adaptativa/genética , Alelos , Animales , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Secuencia de Bases , Evolución Biológica , Población Negra/genética , Regulación de la Expresión Génica , Variación Genética , Humanos , Sistema Inmunológico , Sitios de Carácter Cuantitativo , ARN/genética , Selección Genética , Análisis de Secuencia de ARN , Receptores Toll-Like/genética , Transcripción Genética , Virosis/genética , Virosis/inmunología , Población Blanca/genética
12.
Mol Cell ; 83(3): 481-495, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36334591

RESUMEN

Viral reproduction is contingent on viral protein synthesis that relies on the host ribosomes. As such, viruses have evolved remarkable strategies to hijack the host translational apparatus in order to favor viral protein production and to interfere with cellular innate defenses. Here, we describe the approaches viruses use to exploit the translation machinery, focusing on commonalities across diverse viral families, and discuss the functional relevance of this process. We illustrate the complementary strategies host cells utilize to block viral protein production and consider how cells ensure an efficient antiviral response that relies on translation during this tug of war over the ribosome. Finally, we highlight potential roles mRNA modifications and ribosome quality control play in translational regulation and innate immunity. We address these topics in the context of the COVID-19 pandemic and focus on the gaps in our current knowledge of these mechanisms, specifically in viruses with pandemic potential.


Asunto(s)
COVID-19 , Biosíntesis de Proteínas , Virosis , Virus , Humanos , COVID-19/genética , COVID-19/inmunología , Pandemias , Biosíntesis de Proteínas/genética , Biosíntesis de Proteínas/inmunología , ARN Viral/genética , ARN Viral/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología , Virosis/genética , Virosis/inmunología , Virus/genética , Virus/inmunología , Ribosomas/genética , Ribosomas/inmunología , Ribosomas/virología
13.
Immunity ; 54(4): 753-768.e5, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33765435

RESUMEN

Viral infections induce a conserved host response distinct from bacterial infections. We hypothesized that the conserved response is associated with disease severity and is distinct between patients with different outcomes. To test this, we integrated 4,780 blood transcriptome profiles from patients aged 0 to 90 years infected with one of 16 viruses, including SARS-CoV-2, Ebola, chikungunya, and influenza, across 34 cohorts from 18 countries, and single-cell RNA sequencing profiles of 702,970 immune cells from 289 samples across three cohorts. Severe viral infection was associated with increased hematopoiesis, myelopoiesis, and myeloid-derived suppressor cells. We identified protective and detrimental gene modules that defined distinct trajectories associated with mild versus severe outcomes. The interferon response was decoupled from the protective host response in patients with severe outcomes. These findings were consistent, irrespective of age and virus, and provide insights to accelerate the development of diagnostics and host-directed therapies to improve global pandemic preparedness.


Asunto(s)
Inmunidad/genética , Virosis/inmunología , Presentación de Antígeno/genética , Estudios de Cohortes , Hematopoyesis/genética , Humanos , Interferones/sangre , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Células Mieloides/inmunología , Células Mieloides/patología , Pronóstico , Índice de Severidad de la Enfermedad , Biología de Sistemas , Transcriptoma , Virosis/sangre , Virosis/clasificación , Virosis/genética , Virus/clasificación , Virus/patogenicidad
14.
Nat Immunol ; 18(4): 456-463, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28192417

RESUMEN

Immunodominance (ID) defines the hierarchical immune response to competing antigens in complex immunogens. Little is known regarding B cell and antibody ID despite its importance in immunity to viruses and other pathogens. We show that B cells and serum antibodies from inbred mice demonstrate a reproducible ID hierarchy to the five major antigenic sites in the influenza A virus hemagglutinin globular domain. The hierarchy changed as the immune response progressed, and it was dependent on antigen formulation and delivery. Passive antibody transfer and sequential infection experiments demonstrated 'original antigenic suppression', a phenomenon in which antibodies suppress memory responses to the priming antigenic site. Our study provides a template for attaining deeper understanding of antibody ID to viruses and other complex immunogens.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Interacciones Huésped-Patógeno/inmunología , Epítopos Inmunodominantes/inmunología , Virosis/inmunología , Virus/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Antígenos Virales/química , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antecedentes Genéticos , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Interacciones Huésped-Patógeno/genética , Inmunización , Epítopos Inmunodominantes/química , Memoria Inmunológica , Virus de la Influenza A/inmunología , Ganglios Linfáticos/inmunología , Ratones , Modelos Moleculares , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Conformación Proteica , Virosis/genética , Virosis/virología
15.
Nat Immunol ; 18(7): 744-752, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28553952

RESUMEN

The single-nucleotide polymorphism rs1990760 in the gene encoding the cytosolic viral sensor IFIH1 results in an amino-acid change (A946T; IFIH1T946) that is associated with multiple autoimmune diseases. The effect of this polymorphism on both viral sensing and autoimmune pathogenesis remains poorly understood. Here we found that human peripheral blood mononuclear cells (PBMCs) and cell lines expressing the risk variant IFIH1T946 exhibited heightened basal and ligand-triggered production of type I interferons. Consistent with those findings, mice with a knock-in mutation encoding IFIH1T946 displayed enhanced basal expression of type I interferons, survived a lethal viral challenge and exhibited increased penetrance in autoimmune models, including a combinatorial effect with other risk variants. Furthermore, IFIH1T946 mice manifested an embryonic survival defect consistent with enhanced responsiveness to RNA self ligands. Together our data support a model wherein the production of type I interferons driven by an autoimmune risk variant and triggered by ligand functions to protect against viral challenge, which probably accounts for its selection within human populations but provides this advantage at the cost of modestly promoting the risk of autoimmunity.


Asunto(s)
Autoinmunidad/genética , Infecciones por Cardiovirus/genética , Interferón Tipo I/inmunología , Helicasa Inducida por Interferón IFIH1/genética , Adolescente , Adulto , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Southern Blotting , Infecciones por Cardiovirus/inmunología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Virus de la Encefalomiocarditis/inmunología , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Immunoblotting , Helicasa Inducida por Interferón IFIH1/inmunología , Masculino , Ratones , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virosis/genética , Virosis/inmunología , Adulto Joven
16.
Immunity ; 53(6): 1168-1181.e7, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33326766

RESUMEN

Viruses have evolved multiple strategies to evade elimination by the immune system. Here we examined the contribution of host long noncoding RNAs (lncRNAs) in viral immune evasion. By functional screening of lncRNAs whose expression decreased upon viral infection of macrophages, we identified a lncRNA (lncRNA-GM, Gene Symbol: AK189470.1) that promoted type I interferon (IFN-I) production and inhibited viral replication. Deficiency of lncRNA-GM in mice increased susceptibility to viral infection and impaired IFN-I production. Mechanistically, lncRNA-GM bound to glutathione S-transferase M1 (GSTM1) and blocked GSTM1 interaction with the kinase TBK1, reducing GSTM1-mediated S-glutathionylation of TBK1. Decreased S-glutathionylation enhanced TBK1 activity and downstream production of antiviral mediators. Viral infection reprogrammed intracellular glutathione metabolism and furthermore, an oxidized glutathione mimetic could inhibit TBK1 activity and promote viral replication. Our findings reveal regulation of TBK1 by S-glutathionylation and provide insight into the viral mediated metabolic changes that impact innate immunity and viral evasion.


Asunto(s)
Glutatión/metabolismo , Evasión Inmune , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Glutatión Transferasa/metabolismo , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , ARN Largo no Codificante/genética , Transducción de Señal , Virosis/genética , Virosis/inmunología , Virosis/metabolismo , Replicación Viral
17.
Immunity ; 52(6): 1039-1056.e9, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32392463

RESUMEN

The phenotypic and functional dichotomy between IRF8+ type 1 and IRF4+ type 2 conventional dendritic cells (cDC1s and cDC2s, respectively) is well accepted; it is unknown how robust this dichotomy is under inflammatory conditions, when additionally monocyte-derived cells (MCs) become competent antigen-presenting cells (APCs). Using single-cell technologies in models of respiratory viral infection, we found that lung cDC2s acquired expression of the Fc receptor CD64 shared with MCs and of IRF8 shared with cDC1s. These inflammatory cDC2s (inf-cDC2s) were superior in inducing CD4+ T helper (Th) cell polarization while simultaneously presenting antigen to CD8+ T cells. When carefully separated from inf-cDC2s, MCs lacked APC function. Inf-cDC2s matured in response to cell-intrinsic Toll-like receptor and type 1 interferon receptor signaling, upregulated an IRF8-dependent maturation module, and acquired antigens via convalescent serum and Fc receptors. Because hybrid inf-cDC2s are easily confused with monocyte-derived cells, their existence could explain why APC functions have been attributed to MCs.


Asunto(s)
Plasticidad de la Célula/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunidad , Macrófagos/inmunología , Macrófagos/metabolismo , Infecciones por Respirovirus/etiología , Presentación de Antígeno , Biomarcadores , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Inmunofenotipificación , Interferón Tipo I/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Especificidad de Órganos/inmunología , Receptores Fc/metabolismo , Infecciones por Respirovirus/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Factores de Transcripción , Virosis/genética , Virosis/inmunología , Virosis/metabolismo , Virosis/virología
18.
Nature ; 615(7953): 720-727, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922599

RESUMEN

Engineering the genetic code of an organism has been proposed to provide a firewall from natural ecosystems by preventing viral infections and gene transfer1-6. However, numerous viruses and mobile genetic elements encode parts of the translational apparatus7-9, potentially rendering a genetic-code-based firewall ineffective. Here we show that such mobile transfer RNAs (tRNAs) enable gene transfer and allow viral replication in Escherichia coli despite the genome-wide removal of 3 of the 64 codons and the previously essential cognate tRNA and release factor genes. We then establish a genetic firewall by discovering viral tRNAs that provide exceptionally efficient codon reassignment allowing us to develop cells bearing an amino acid-swapped genetic code that reassigns two of the six serine codons to leucine during translation. This amino acid-swapped genetic code renders cells resistant to viral infections by mistranslating viral proteomes and prevents the escape of synthetic genetic information by engineered reliance on serine codons to produce leucine-requiring proteins. As these cells may have a selective advantage over wild organisms due to virus resistance, we also repurpose a third codon to biocontain this virus-resistant host through dependence on an amino acid not found in nature10. Our results may provide the basis for a general strategy to make any organism safely resistant to all natural viruses and prevent genetic information flow into and out of genetically modified organisms.


Asunto(s)
Aminoácidos , Escherichia coli , Transferencia de Gen Horizontal , Código Genético , Interacciones Microbiota-Huesped , Biosíntesis de Proteínas , Virosis , Aminoácidos/genética , Aminoácidos/metabolismo , Codón/genética , Ecosistema , Escherichia coli/genética , Escherichia coli/virología , Código Genético/genética , Leucina/genética , Leucina/metabolismo , Biosíntesis de Proteínas/genética , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Serina/genética , Virosis/genética , Virosis/prevención & control , Interacciones Microbiota-Huesped/genética , Organismos Modificados Genéticamente/genética , Genoma Bacteriano/genética , Transferencia de Gen Horizontal/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
19.
Immunity ; 51(5): 840-855.e5, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31606264

RESUMEN

TCF-1 is a key transcription factor in progenitor exhausted CD8 T cells (Tex). Moreover, this Tex cell subset mediates responses to PD-1 checkpoint pathway blockade. However, the role of the transcription factor TCF-1 in early fate decisions and initial generation of Tex cells is unclear. Single-cell RNA sequencing (scRNA-seq) and lineage tracing identified a TCF-1+Ly108+PD-1+ CD8 T cell population that seeds development of mature Tex cells early during chronic infection. TCF-1 mediated the bifurcation between divergent fates, repressing development of terminal KLRG1Hi effectors while fostering KLRG1Lo Tex precursor cells, and PD-1 stabilized this TCF-1+ Tex precursor cell pool. TCF-1 mediated a T-bet-to-Eomes transcription factor transition in Tex precursors by promoting Eomes expression and drove c-Myb expression that controlled Bcl-2 and survival. These data define a role for TCF-1 in early-fate-bifurcation-driving Tex precursor cells and also identify PD-1 as a protector of this early TCF-1 subset.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Redes Reguladoras de Genes , Factor 1 de Transcripción de Linfocitos T/metabolismo , Transcripción Genética , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Enfermedad Crónica , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Ratones , Receptor de Muerte Celular Programada 1/metabolismo , Factor 1 de Transcripción de Linfocitos T/genética , Virosis/genética , Virosis/inmunología , Virosis/virología
20.
Immunity ; 51(5): 856-870.e5, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31747582

RESUMEN

Naive CD8+ T cells differentiating into effector T cells increase glucose uptake and shift from quiescent to anabolic metabolism. Although much is known about the metabolism of cultured T cells, how T cells use nutrients during immune responses in vivo is less well defined. Here, we combined bioenergetic profiling and 13C-glucose infusion techniques to investigate the metabolism of CD8+ T cells responding to Listeria infection. In contrast to in vitro-activated T cells, which display hallmarks of Warburg metabolism, physiologically activated CD8+ T cells displayed greater rates of oxidative metabolism, higher bioenergetic capacity, differential use of pyruvate, and prominent flow of 13C-glucose carbon to anabolic pathways, including nucleotide and serine biosynthesis. Glucose-dependent serine biosynthesis mediated by the enzyme Phgdh was essential for CD8+ T cell expansion in vivo. Our data highlight fundamental differences in glucose use by pathogen-specific T cells in vivo, illustrating the impact of environment on T cell metabolic phenotypes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Activación de Linfocitos/inmunología , Metaboloma , Metabolómica , Animales , Proliferación Celular , Cromatografía de Gases y Espectrometría de Masas , Glucólisis , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Activación de Linfocitos/genética , Metabolómica/métodos , Ratones , Estrés Oxidativo , Virosis/genética , Virosis/inmunología , Virosis/metabolismo , Virosis/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA