Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 274.359
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 42(1): 347-373, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38941603

RESUMEN

Plasmacytoid dendritic cells (pDCs) represent a unique cell type within the innate immune system. Their defining property is the recognition of pathogen-derived nucleic acids through endosomal Toll-like receptors and the ensuing production of type I interferon and other soluble mediators, which orchestrate innate and adaptive responses. We review several aspects of pDC biology that have recently come to the fore. We discuss emerging questions regarding the lineage affiliation and origin of pDCs and argue that these cells constitute an integral part of the dendritic cell lineage. We emphasize the specific function of pDCs as innate sentinels of virus infection, particularly their recognition of and distinct response to virus-infected cells. This essential evolutionary role of pDCs has been particularly important for the control of coronaviruses, as demonstrated by the recent COVID-19 pandemic. Finally, we highlight the key contribution of pDCs to systemic lupus erythematosus, in which therapeutic targeting of pDCs is currently underway.


Asunto(s)
COVID-19 , Células Dendríticas , Inmunidad Innata , Lupus Eritematoso Sistémico , SARS-CoV-2 , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , COVID-19/inmunología , Animales , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Lupus Eritematoso Sistémico/inmunología , Receptores Toll-Like/metabolismo , Diferenciación Celular , Linaje de la Célula
2.
Annu Rev Immunol ; 42(1): 615-645, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38941608

RESUMEN

The COVID-19 pandemic was caused by the recently emerged ß-coronavirus SARS-CoV-2. SARS-CoV-2 has had a catastrophic impact, resulting in nearly 7 million fatalities worldwide to date. The innate immune system is the first line of defense against infections, including the detection and response to SARS-CoV-2. Here, we discuss the innate immune mechanisms that sense coronaviruses, with a focus on SARS-CoV-2 infection and how these protective responses can become detrimental in severe cases of COVID-19, contributing to cytokine storm, inflammation, long-COVID, and other complications. We also highlight the complex cross talk among cytokines and the cellular components of the innate immune system, which can aid in viral clearance but also contribute to inflammatory cell death, cytokine storm, and organ damage in severe COVID-19 pathogenesis. Furthermore, we discuss how SARS-CoV-2 evades key protective innate immune mechanisms to enhance its virulence and pathogenicity, as well as how innate immunity can be therapeutically targeted as part of the vaccination and treatment strategy. Overall, we highlight how a comprehensive understanding of innate immune mechanisms has been crucial in the fight against SARS-CoV-2 infections and the development of novel host-directed immunotherapeutic strategies for various diseases.


Asunto(s)
COVID-19 , Inmunidad Innata , SARS-CoV-2 , Humanos , COVID-19/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Síndrome de Liberación de Citoquinas/inmunología , Citocinas/metabolismo , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/prevención & control , Evasión Inmune
3.
Annu Rev Immunol ; 41: 277-300, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-36716750

RESUMEN

Emerging and re-emerging respiratory viral infections pose a tremendous threat to human society, as exemplified by the ongoing COVID-19 pandemic. Upon viral invasion of the respiratory tract, the host initiates coordinated innate and adaptive immune responses to defend against the virus and to promote repair of the damaged tissue. However, dysregulated host immunity can also cause acute morbidity, hamper lung regeneration, and/or lead to chronic tissue sequelae. Here, we review our current knowledge of the immune mechanisms regulating antiviral protection, host pathogenesis, inflammation resolution, and lung regeneration following respiratory viral infections, mainly using influenza virus and SARS-CoV-2 infections as examples. We hope that this review sheds light on future research directions to elucidate the cellular and molecular cross talk regulating host recovery and to pave the way to the development of pro-repair therapeutics to augment lung regeneration following viral injury.


Asunto(s)
COVID-19 , Humanos , Animales , Inmunidad Innata , Pandemias , SARS-CoV-2 , Inflamación/patología
4.
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
5.
Annu Rev Immunol ; 41: 561-585, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37126418

RESUMEN

Infection with SARS-CoV-2 results in clinical outcomes ranging from silent or benign infection in most individuals to critical pneumonia and death in a few. Genetic studies in patients have established that critical cases can result from inborn errors of TLR3- or TLR7-dependent type I interferon immunity, or from preexisting autoantibodies neutralizing primarily IFN-α and/or IFN-ω. These findings are consistent with virological studies showing that multiple SARS-CoV-2 proteins interfere with pathways of induction of, or response to, type I interferons. They are also congruent with cellular studies and mouse models that found that type I interferons can limit SARS-CoV-2 replication in vitro and in vivo, while their absence or diminution unleashes viral growth. Collectively, these findings point to insufficient type I interferon during the first days of infection as a general mechanism underlying critical COVID-19 pneumonia, with implications for treatment and directions for future research.


Asunto(s)
COVID-19 , Interferón Tipo I , Ratones , Humanos , Animales , Interferones/farmacología , SARS-CoV-2
6.
Annu Rev Immunol ; 40: 349-386, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35113730

RESUMEN

Antibodies have been used to prevent or treat viral infections since the nineteenth century, but the full potential to use passive immunization for infectious diseases has yet to be realized. The advent of efficient methods for isolating broad and potently neutralizing human monoclonal antibodies is enabling us to develop antibodies with unprecedented activities. The discovery of IgG Fc region modifications that extend antibody half-life in humans to three months or more suggests that antibodies could become the principal tool with which we manage future viral epidemics. Antibodies for members of most virus families that cause severe disease in humans have been isolated, and many of them are in clinical development, an area that has accelerated during the effort to prevent or treat COVID-19 (coronavirus disease 2019). Broad and potently neutralizing antibodies are also important research reagents for identification of protective epitopes that can be engineered into active vaccines through structure-based reverse vaccinology.


Asunto(s)
Anticuerpos Antivirales , COVID-19 , Animales , Anticuerpos Neutralizantes , Epítopos , Humanos , Inmunización Pasiva/métodos
7.
Annu Rev Immunol ; 40: 323-348, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35113729

RESUMEN

The diverse biological activity of interleukin-6 (IL-6) contributes to the maintenance of homeostasis. Emergent infection or tissue injury induces rapid production of IL-6 and activates host defense through augmentation of acute-phase proteins and immune responses. However, excessive IL-6 production and uncontrolled IL-6 receptor signaling are critical to pathogenesis. Over the years, therapeutic agents targeting IL-6 signaling, such as tocilizumab, a humanized anti-IL-6 receptor antibody, have shown remarkable efficacy for rheumatoid arthritis, Castleman disease, and juvenile idiopathic arthritis, and their efficacy in other diseases is continually being reported. Emerging evidence has demonstrated the benefit of tocilizumab for several types of acute inflammatory diseases, including cytokine storms induced by chimeric antigen receptor T cell therapy and coronavirus disease 2019 (COVID-19). Here, we refocus attention on the biology of IL-6 and summarize the distinct pathological roles of IL-6 signaling in several acute and chronic inflammatory diseases.


Asunto(s)
Artritis Reumatoide , COVID-19 , Animales , Artritis Reumatoide/terapia , COVID-19/terapia , Humanos , Inmunoterapia Adoptiva , Interleucina-6/metabolismo , Transducción de Señal
8.
Annu Rev Immunol ; 40: 443-467, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35471837

RESUMEN

A principal purpose of type 2 immunity was thought to be defense against large parasites, but it also functions in the restoration of homeostasis, such as toxin clearance following snake bites. In other cases, like allergy, the type 2 T helper (Th2) cytokines and cells present in the environment are detrimental and cause diseases. In recent years, the recognition of cell heterogeneity within Th2-associated cell populations has revealed specific functions of cells with a particular phenotype or gene signature. In addition, here we discuss the recent data regarding heterogeneity of type 2 immunity-related cells, as well as their newly identified role in a variety of processes ranging from involvement in respiratory viral infections [especially in the context of the recent COVID-19 (coronavirus disease 2019) pandemic] to control of cancer development or of metabolic homeostasis.


Asunto(s)
COVID-19 , Hipersensibilidad , Animales , Citocinas/metabolismo , Homeostasis , Humanos , Linfocitos T Colaboradores-Inductores/metabolismo , Células Th2
9.
Annu Rev Immunol ; 40: 75-94, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-34985929

RESUMEN

Strong epidemiological evidence now exists that sex is an important biologic variable in immunity. Recent studies, for example, have revealed that sex differences are associated with the severity of symptoms and mortality due to coronavirus disease 2019 (COVID-19). Despite this evidence, much remains to be learned about the mechanisms underlying associations between sex differences and immune-mediated conditions. A growing body of experimental data has made significant inroads into understanding sex-influenced immune responses. As physicians seek to provide more targeted patient care, it is critical to understand how sex-defining factors (e.g., chromosomes, gonadal hormones) alter immune responses in health and disease. In this review, we highlight recent insights into sex differences in autoimmunity; virus infection, specifically severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection; and cancer immunotherapy. A deeper understanding of underlying mechanisms will allow the development of a sex-based approach to disease screening and treatment.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Femenino , Humanos , Masculino , Caracteres Sexuales , Factores Sexuales
10.
Cell ; 187(3): 585-595.e6, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38194968

RESUMEN

Evolution of SARS-CoV-2 requires the reassessment of current vaccine measures. Here, we characterized BA.2.86 and XBB-derived variant FLip by investigating their neutralization alongside D614G, BA.1, BA.2, BA.4/5, XBB.1.5, and EG.5.1 by sera from 3-dose-vaccinated and bivalent-vaccinated healthcare workers, XBB.1.5-wave-infected first responders, and monoclonal antibody (mAb) S309. We assessed the biology of the variant spikes by measuring viral infectivity and membrane fusogenicity. BA.2.86 is less immune evasive compared to FLip and other XBB variants, consistent with antigenic distances. Importantly, distinct from XBB variants, mAb S309 was unable to neutralize BA.2.86, likely due to a D339H mutation based on modeling. BA.2.86 had relatively high fusogenicity and infectivity in CaLu-3 cells but low fusion and infectivity in 293T-ACE2 cells compared to some XBB variants, suggesting a potentially different conformational stability of BA.2.86 spike. Overall, our study underscores the importance of SARS-CoV-2 variant surveillance and the need for updated COVID-19 vaccines.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Evasión Inmune , SARS-CoV-2 , Humanos , Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/inmunología , SARS-CoV-2/clasificación , SARS-CoV-2/fisiología
11.
Cell ; 187(8): 1822-1822.e1, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38608649

RESUMEN

In November 2023, Japan's Ministry of Health, Labour and Welfare granted regulatory approval of ARCT-154, a self-amplifying RNA COVID-19 vaccine developed by Arcturus Therapeutics. Clinical trials showed comparable safety and efficacy using a lower dose compared to the mRNA vaccine BNT162b2. To view this Bench-to-Bedside, open or download the PDF.


Asunto(s)
Vacunas contra la COVID-19 , Vacunas de ARNm , Humanos , Vacuna BNT162 , COVID-19/prevención & control , Vacunas de ARNm/administración & dosificación , Vacunas de ARNm/efectos adversos
12.
Cell ; 187(10): 2393-2410.e14, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38653235

RESUMEN

SARS-CoV-2 and other sarbecoviruses continue to threaten humanity, highlighting the need to characterize common mechanisms of viral immune evasion for pandemic preparedness. Cytotoxic lymphocytes are vital for antiviral immunity and express NKG2D, an activating receptor conserved among mammals that recognizes infection-induced stress ligands (e.g., MIC-A/B). We found that SARS-CoV-2 evades NKG2D recognition by surface downregulation of MIC-A/B via shedding, observed in human lung tissue and COVID-19 patient serum. Systematic testing of SARS-CoV-2 proteins revealed that ORF6, an accessory protein uniquely conserved among sarbecoviruses, was responsible for MIC-A/B downregulation via shedding. Further investigation demonstrated that natural killer (NK) cells efficiently killed SARS-CoV-2-infected cells and limited viral spread. However, inhibition of MIC-A/B shedding with a monoclonal antibody, 7C6, further enhanced NK-cell activity toward SARS-CoV-2-infected cells. Our findings unveil a strategy employed by SARS-CoV-2 to evade cytotoxic immunity, identify the culprit immunevasin shared among sarbecoviruses, and suggest a potential novel antiviral immunotherapy.


Asunto(s)
COVID-19 , Evasión Inmune , Células Asesinas Naturales , Subfamilia K de Receptores Similares a Lectina de Células NK , SARS-CoV-2 , Humanos , SARS-CoV-2/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , COVID-19/inmunología , COVID-19/virología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Animales , Citotoxicidad Inmunológica , Regulación hacia Abajo , Pulmón/inmunología , Pulmón/virología , Pulmón/patología
13.
Cell ; 187(3): 596-608.e17, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38194966

RESUMEN

BA.2.86, a recently identified descendant of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron BA.2 sublineage, contains ∼35 mutations in the spike (S) protein and spreads in multiple countries. Here, we investigated whether the virus exhibits altered biological traits, focusing on S protein-driven viral entry. Employing pseudotyped particles, we show that BA.2.86, unlike other Omicron sublineages, enters Calu-3 lung cells with high efficiency and in a serine- but not cysteine-protease-dependent manner. Robust lung cell infection was confirmed with authentic BA.2.86, but the virus exhibited low specific infectivity. Further, BA.2.86 was highly resistant against all therapeutic antibodies tested, efficiently evading neutralization by antibodies induced by non-adapted vaccines. In contrast, BA.2.86 and the currently circulating EG.5.1 sublineage were appreciably neutralized by antibodies induced by the XBB.1.5-adapted vaccine. Collectively, BA.2.86 has regained a trait characteristic of early SARS-CoV-2 lineages, robust lung cell entry, and evades neutralizing antibodies. However, BA.2.86 exhibits low specific infectivity, which might limit transmissibility.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19 , SARS-CoV-2 , Humanos , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Caspasas/metabolismo , COVID-19/inmunología , COVID-19/virología , Pulmón/virología , SARS-CoV-2/clasificación , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Internalización del Virus , Glicoproteína de la Espiga del Coronavirus/genética
15.
Cell ; 186(18): 3753-3755, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37657416

RESUMEN

In addition to acute hyperinflammatory responses, SARS-CoV-2 infections can have long-term effects on our immune system leading to, for example, post-acute sequelae of COVID-19 (PASC). In this issue of Cell, Cheong et al. show that severe infections via IL-6 induce persistent epigenetic signatures in hemopoietic stem cells and their myeloid progenitors associated with increased inflammatory potential.


Asunto(s)
COVID-19 , Síndrome Post Agudo de COVID-19 , Humanos , COVID-19/genética , COVID-19/inmunología , COVID-19/patología , Epigenómica , Células Madre Hematopoyéticas , Síndrome Post Agudo de COVID-19/inmunología , Síndrome Post Agudo de COVID-19/patología , SARS-CoV-2 , Inmunidad Entrenada , Inflamación/patología
16.
Cell ; 186(2): 279-286.e8, 2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36580913

RESUMEN

The BQ and XBB subvariants of SARS-CoV-2 Omicron are now rapidly expanding, possibly due to altered antibody evasion properties deriving from their additional spike mutations. Here, we report that neutralization of BQ.1, BQ.1.1, XBB, and XBB.1 by sera from vaccinees and infected persons was markedly impaired, including sera from individuals boosted with a WA1/BA.5 bivalent mRNA vaccine. Titers against BQ and XBB subvariants were lower by 13- to 81-fold and 66- to 155-fold, respectively, far beyond what had been observed to date. Monoclonal antibodies capable of neutralizing the original Omicron variant were largely inactive against these new subvariants, and the responsible individual spike mutations were identified. These subvariants were found to have similar ACE2-binding affinities as their predecessors. Together, our findings indicate that BQ and XBB subvariants present serious threats to current COVID-19 vaccines, render inactive all authorized antibodies, and may have gained dominance in the population because of their advantage in evading antibodies.


Asunto(s)
Anticuerpos Antivirales , COVID-19 , Evasión Inmune , SARS-CoV-2 , Humanos , Anticuerpos Monoclonales , Anticuerpos Neutralizantes , COVID-19/inmunología , COVID-19/virología , Vacunas contra la COVID-19 , SARS-CoV-2/clasificación , SARS-CoV-2/genética
17.
Cell ; 186(23): 5151-5164.e13, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37875109

RESUMEN

The large-scale evolution of the SARS-CoV-2 virus has been marked by rapid turnover of genetic clades. New variants show intrinsic changes, notably increased transmissibility, and antigenic changes that reduce cross-immunity induced by previous infections or vaccinations. How this functional variation shapes global evolution has remained unclear. Here, we establish a predictive fitness model for SARS-CoV-2 that integrates antigenic and intrinsic selection. The model is informed by tracking of time-resolved sequence data, epidemiological records, and cross-neutralization data of viral variants. Our inference shows that immune pressure, including contributions of vaccinations and previous infections, has become the dominant force driving the recent evolution of SARS-CoV-2. The fitness model can serve continued surveillance in two ways. First, it successfully predicts the short-term evolution of circulating strains and flags emerging variants likely to displace the previously predominant variant. Second, it predicts likely antigenic profiles of successful escape variants prior to their emergence.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/epidemiología , COVID-19/virología , SARS-CoV-2/genética , SARS-CoV-2/fisiología , Vacunación , Modelos Genéticos , Monitoreo Epidemiológico
18.
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
19.
Cell ; 186(11): 2392-2409.e21, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-37164012

RESUMEN

T cell responses play an important role in protection against beta-coronavirus infections, including SARS-CoV-2, where they associate with decreased COVID-19 disease severity and duration. To enhance T cell immunity across epitopes infrequently altered in SARS-CoV-2 variants, we designed BNT162b4, an mRNA vaccine component that is intended to be combined with BNT162b2, the spike-protein-encoding vaccine. BNT162b4 encodes variant-conserved, immunogenic segments of the SARS-CoV-2 nucleocapsid, membrane, and ORF1ab proteins, targeting diverse HLA alleles. BNT162b4 elicits polyfunctional CD4+ and CD8+ T cell responses to diverse epitopes in animal models, alone or when co-administered with BNT162b2 while preserving spike-specific immunity. Importantly, we demonstrate that BNT162b4 protects hamsters from severe disease and reduces viral titers following challenge with viral variants. These data suggest that a combination of BNT162b2 and BNT162b4 could reduce COVID-19 disease severity and duration caused by circulating or future variants. BNT162b4 is currently being clinically evaluated in combination with the BA.4/BA.5 Omicron-updated bivalent BNT162b2 (NCT05541861).


Asunto(s)
Vacuna BNT162 , COVID-19 , Animales , Cricetinae , Humanos , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Epítopos , SARS-CoV-2/genética
20.
Cell ; 186(21): 4597-4614.e26, 2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37738970

RESUMEN

SARS-CoV-2 variants of concern (VOCs) emerged during the COVID-19 pandemic. Here, we used unbiased systems approaches to study the host-selective forces driving VOC evolution. We discovered that VOCs evolved convergent strategies to remodel the host by modulating viral RNA and protein levels, altering viral and host protein phosphorylation, and rewiring virus-host protein-protein interactions. Integrative computational analyses revealed that although Alpha, Beta, Gamma, and Delta ultimately converged to suppress interferon-stimulated genes (ISGs), Omicron BA.1 did not. ISG suppression correlated with the expression of viral innate immune antagonist proteins, including Orf6, N, and Orf9b, which we mapped to specific mutations. Later Omicron subvariants BA.4 and BA.5 more potently suppressed innate immunity than early subvariant BA.1, which correlated with Orf6 levels, although muted in BA.4 by a mutation that disrupts the Orf6-nuclear pore interaction. Our findings suggest that SARS-CoV-2 convergent evolution overcame human adaptive and innate immune barriers, laying the groundwork to tackle future pandemics.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/virología , Inmunidad Innata/genética , Pandemias , SARS-CoV-2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA