Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.386
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Annu Rev Immunol ; 36: 279-308, 2018 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-29345964

RESUMO

Flaviviruses such as dengue (DENV), yellow fever (YFV), West Nile (WNV), and Zika (ZIKV) are human pathogens of global significance. In particular, DENV causes the most prevalent mosquito-borne viral diseases in humans, and ZIKV emerged from obscurity into the spotlight in 2016 as the etiologic agent of congenital Zika syndrome. Owing to the recent emergence of ZIKV as a global pandemic threat, the roles of the immune system during ZIKV infections are as yet unclear. In contrast, decades of DENV research implicate a dual role for the immune system in protection against and pathogenesis of DENV infection. As DENV and ZIKV are closely related, knowledge based on DENV studies has been used to prioritize investigation of ZIKV immunity and pathogenesis, and to accelerate ZIKV diagnostic, therapeutic, and vaccine design. This review discusses the following topics related to innate and adaptive immune responses to DENV and ZIKV: the interferon system as the key mechanism of host defense and viral target for immune evasion, antibody-mediated protection versus antibody-dependent enhancement, and T cell-mediated protection versus original T cell antigenic sin. Understanding the mechanisms that regulate the balance between immune-mediated protection and pathogenesis during DENV and ZIKV infections is critical toward development of safe and effective DENV and ZIKV therapeutics and vaccines.


Assuntos
Vírus da Dengue/fisiologia , Dengue/imunologia , Interações Hospedeiro-Patógeno/imunologia , Infecção por Zika virus/imunologia , Zika virus/fisiologia , Imunidade Adaptativa , Animais , Dengue/metabolismo , Dengue/prevenção & controle , Dengue/virologia , Humanos , Imunidade Inata , Interferon Tipo I/metabolismo , Tropismo Viral , Vacinas Virais/imunologia , Infecção por Zika virus/metabolismo , Infecção por Zika virus/prevenção & controle , Infecção por Zika virus/virologia
2.
Cell ; 184(25): 6052-6066.e18, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34852239

RESUMO

The human monoclonal antibody C10 exhibits extraordinary cross-reactivity, potently neutralizing Zika virus (ZIKV) and the four serotypes of dengue virus (DENV1-DENV4). Here we describe a comparative structure-function analysis of C10 bound to the envelope (E) protein dimers of the five viruses it neutralizes. We demonstrate that the C10 Fab has high affinity for ZIKV and DENV1 but not for DENV2, DENV3, and DENV4. We further show that the C10 interaction with the latter viruses requires an E protein conformational landscape that limits binding to only one of the three independent epitopes per virion. This limited affinity is nevertheless counterbalanced by the particle's icosahedral organization, which allows two different dimers to be reached by both Fab arms of a C10 immunoglobulin. The epitopes' geometric distribution thus confers C10 its exceptional neutralization breadth. Our results highlight the importance not only of paratope/epitope complementarity but also the topological distribution for epitope-focused vaccine design.


Assuntos
Anticorpos Neutralizantes , Vírus da Dengue , Dengue , Proteínas do Envelope Viral , Infecção por Zika virus , Zika virus , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/imunologia , Linhagem Celular , Chlorocebus aethiops , Reações Cruzadas/imunologia , Dengue/imunologia , Dengue/virologia , Vírus da Dengue/imunologia , Vírus da Dengue/fisiologia , Drosophila melanogaster , Células HEK293 , Humanos , Ligação Proteica , Conformação Proteica , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo , Zika virus/imunologia , Zika virus/fisiologia , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia
3.
Cell ; 176(4): 687-701.e5, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30735632

RESUMO

Female Aedes aegypti mosquitoes bite humans to obtain blood to develop their eggs. Remarkably, their strong attraction to humans is suppressed for days after the blood meal by an unknown mechanism. We investigated a role for neuropeptide Y (NPY)-related signaling in long-term behavioral suppression and discovered that drugs targeting human NPY receptors modulate mosquito host-seeking. In a screen of all 49 predicted Ae. aegypti peptide receptors, we identified NPY-like receptor 7 (NPYLR7) as the sole target of these drugs. To obtain small-molecule agonists selective for NPYLR7, we performed a high-throughput cell-based assay of 265,211 compounds and isolated six highly selective NPYLR7 agonists that inhibit mosquito attraction to humans. NPYLR7 CRISPR-Cas9 null mutants are defective in behavioral suppression and resistant to these drugs. Finally, we show that these drugs can inhibit biting and blood-feeding on a live host, suggesting a novel approach to control infectious disease transmission by controlling mosquito behavior. VIDEO ABSTRACT.


Assuntos
Comportamento de Busca por Hospedeiro/efeitos dos fármacos , Mosquitos Vetores/efeitos dos fármacos , Receptores de Neuropeptídeo Y/agonistas , Aedes/metabolismo , Animais , Comportamento Alimentar/efeitos dos fármacos , Feminino , Células HEK293 , Humanos , Mordeduras e Picadas de Insetos , Receptores de Neuropeptídeo Y/metabolismo , Bibliotecas de Moléculas Pequenas/análise
4.
Cell ; 175(7): 1931-1945.e18, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30550790

RESUMO

Mosquito-borne flaviviruses, including dengue virus (DENV) and Zika virus (ZIKV), are a growing public health concern. Systems-level analysis of how flaviviruses hijack cellular processes through virus-host protein-protein interactions (PPIs) provides information about their replication and pathogenic mechanisms. We used affinity purification-mass spectrometry (AP-MS) to compare flavivirus-host interactions for two viruses (DENV and ZIKV) in two hosts (human and mosquito). Conserved virus-host PPIs revealed that the flavivirus NS5 protein suppresses interferon stimulated genes by inhibiting recruitment of the transcription complex PAF1C and that chemical modulation of SEC61 inhibits DENV and ZIKV replication in human and mosquito cells. Finally, we identified a ZIKV-specific interaction between NS4A and ANKLE2, a gene linked to hereditary microcephaly, and showed that ZIKV NS4A causes microcephaly in Drosophila in an ANKLE2-dependent manner. Thus, comparative flavivirus-host PPI mapping provides biological insights and, when coupled with in vivo models, can be used to unravel pathogenic mechanisms.


Assuntos
Vírus da Dengue , Dengue , Proteínas de Membrana , Proteínas Nucleares , Proteínas não Estruturais Virais , Infecção por Zika virus , Zika virus , Animais , Linhagem Celular Tumoral , Culicidae , Dengue/genética , Dengue/metabolismo , Dengue/patologia , Vírus da Dengue/genética , Vírus da Dengue/metabolismo , Vírus da Dengue/patogenicidade , Células HEK293 , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Mapeamento de Interação de Proteínas , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Zika virus/genética , Zika virus/metabolismo , Zika virus/patogenicidade , Infecção por Zika virus/genética , Infecção por Zika virus/metabolismo , Infecção por Zika virus/patologia
5.
Cell ; 169(4): 597-609.e11, 2017 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-28475892

RESUMO

Antibodies to Zika virus (ZIKV) can be protective. To examine the antibody response in individuals who develop high titers of anti-ZIKV antibodies, we screened cohorts in Brazil and Mexico for ZIKV envelope domain III (ZEDIII) binding and neutralization. We find that serologic reactivity to dengue 1 virus (DENV1) EDIII before ZIKV exposure is associated with increased ZIKV neutralizing titers after exposure. Antibody cloning shows that donors with high ZIKV neutralizing antibody titers have expanded clones of memory B cells that express the same immunoglobulin VH3-23/VK1-5 genes. These recurring antibodies cross-react with DENV1, but not other flaviviruses, neutralize both DENV1 and ZIKV, and protect mice against ZIKV challenge. Structural analyses reveal the mechanism of recognition of the ZEDIII lateral ridge by VH3-23/VK1-5 antibodies. Serologic testing shows that antibodies to this region correlate with serum neutralizing activity to ZIKV. Thus, high neutralizing responses to ZIKV are associated with pre-existing reactivity to DENV1 in humans.


Assuntos
Anticorpos Neutralizantes/química , Anticorpos Antivirais/química , Infecção por Zika virus/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Brasil , Feminino , Humanos , Memória Imunológica , Leucócitos Mononucleares/imunologia , Masculino , México , Camundongos , Infecção por Zika virus/sangue
6.
Cell ; 168(6): 1114-1125.e10, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28222903

RESUMO

The emergence of ZIKV infection has prompted a global effort to develop safe and effective vaccines. We engineered a lipid nanoparticle (LNP) encapsulated modified mRNA vaccine encoding wild-type or variant ZIKV structural genes and tested immunogenicity and protection in mice. Two doses of modified mRNA LNPs encoding prM-E genes that produced virus-like particles resulted in high neutralizing antibody titers (∼1/100,000) that protected against ZIKV infection and conferred sterilizing immunity. To offset a theoretical concern of ZIKV vaccines inducing antibodies that cross-react with the related dengue virus (DENV), we designed modified prM-E RNA encoding mutations destroying the conserved fusion-loop epitope in the E protein. This variant protected against ZIKV and diminished production of antibodies enhancing DENV infection in cells or mice. A modified mRNA vaccine can prevent ZIKV disease and be adapted to reduce the risk of sensitizing individuals to subsequent exposure to DENV, should this become a clinically relevant concern.


Assuntos
RNA Mensageiro/administração & dosagem , Vacinas Virais/imunologia , Infecção por Zika virus/imunologia , Infecção por Zika virus/prevenção & controle , Animais , Epitopos/imunologia , Feminino , Lipídeos/química , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanopartículas/química , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Vacinas Virais/administração & dosagem , Zika virus/imunologia
7.
Mol Cell ; 83(15): 2781-2791.e4, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37478848

RESUMO

Dengue is a mosquito-borne viral infection caused by dengue virus (DENV), a member of the flaviviruses. The DENV genome is a 5'-capped positive-sense RNA with a unique 5'-stem-loop structure (SLA), which is essential for RNA replication and 5' capping. The virus-encoded proteins NS5 and NS3 are responsible for viral genome replication, but the structural basis by which they cooperatively conduct the required tasks has remained unclear. Here, we report the cryoelectron microscopy (cryo-EM) structures of SLA-bound NS5 (PC), NS3-bound PC (PC-NS3), and an RNA-elongating NS5-NS3 complex (EC). While SLA bridges the NS5 methyltransferase and RNA-dependent RNA polymerase domains in PC, the NS3 helicase domain displaces it in elongation complex (EC). The SLA- and NS3-binding sites overlap with that of human STAT2. These structures illuminate the key steps in DENV genome replication, namely, SLA-dependent replication initiation, processive RNA elongation, and 5' capping of the nascent genomic RNA, thereby providing foundations to combat flaviviruses.


Assuntos
Vírus da Dengue , Animais , Humanos , Vírus da Dengue/genética , Microscopia Crioeletrônica , Sítios de Ligação , RNA Polimerase Dependente de RNA/metabolismo , Capuzes de RNA , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , RNA Viral/metabolismo
8.
Immunity ; 53(5): 1078-1094.e7, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33010224

RESUMO

Memory B cells (MBCs) can respond to heterologous antigens either by molding new specificities through secondary germinal centers (GCs) or by selecting preexisting clones without further affinity maturation. To distinguish these mechanisms in flavivirus infections and immunizations, we studied recall responses to envelope protein domain III (DIII). Conditional deletion of activation-induced cytidine deaminase (AID) between heterologous challenges of West Nile, Japanese encephalitis, Zika, and dengue viruses did not affect recall responses. DIII-specific MBCs were contained mostly within the plasma-cell-biased CD80+ subset, and few GCs arose following heterologous boosters, demonstrating that recall responses are confined by preexisting clonal diversity. Measurement of monoclonal antibody (mAb) binding affinity to DIII proteins, timed AID deletion, single-cell RNA sequencing, and lineage tracing experiments point to selection of relatively low-affinity MBCs as a mechanism to promote diversity. Engineering immunogens to avoid this MBC diversity may facilitate flavivirus-type-specific vaccines with minimized potential for infection enhancement.


Assuntos
Linfócitos B/imunologia , Reações Cruzadas/imunologia , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/virologia , Flavivirus/imunologia , Interações Hospedeiro-Patógeno/imunologia , Memória Imunológica , Animais , Linfócitos B/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta Imunológica , Infecções por Flavivirus/metabolismo , Imunização , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Plasmócitos/imunologia , Plasmócitos/metabolismo , Especificidade da Espécie
9.
Immunity ; 51(6): 1119-1135.e5, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31757672

RESUMO

T cells play important multifaceted roles during dengue infection, and understanding their responses is important for defining correlates of protective immunity and identifying effective vaccine antigens. Using mass cytometry and a highly multiplexed peptide-HLA (human leukocyte antigen) tetramer staining strategy, we probed T cells from dengue patients-a total of 430 dengue and control candidate epitopes-together with key markers of activation, trafficking, and differentiation. During acute disease, dengue-specific CD8+ T cells expressed a distinct profile of activation and trafficking receptors that distinguished them from non-dengue-specific T cells. During convalescence, dengue-specific T cells differentiated into two major cell fates, CD57+ CD127--resembling terminally differentiated senescent memory cells and CD127+ CD57--resembling proliferation-capable memory cells. Validation in an independent cohort showed that these subsets remained at elevated frequencies up to one year after infection. These analyses aid our understanding of the generation of T cell memory in dengue infection or vaccination.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vírus da Dengue/imunologia , Dengue/imunologia , Antígenos HLA/imunologia , Adulto , Linfócitos B/imunologia , Antígenos CD57/metabolismo , Diferenciação Celular/imunologia , Proliferação de Células/fisiologia , Epitopos de Linfócito T/imunologia , Feminino , Antígenos HLA/classificação , Humanos , Memória Imunológica/imunologia , Subunidade alfa de Receptor de Interleucina-7/metabolismo , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Masculino , Pessoa de Meia-Idade
10.
Immunity ; 50(3): 751-762.e5, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30737148

RESUMO

Zika virus (ZIKV) has recently been associated with birth defects and pregnancy loss after maternal infection. Because dengue virus (DENV) and ZIKV co-circulate, understanding the role of antibody-dependent enhancement in the context of pregnancy is critical. Here, we showed that the presence of DENV-specific antibodies in ZIKV-infected pregnant mice significantly increased placental damage, fetal growth restriction, and fetal resorption. This was associated with enhanced viral replication in the placenta that coincided with an increased frequency of infected trophoblasts. ZIKV-infected human placental tissues also showed increased replication in the presence of DENV antibodies, which was reversed by FcγR blocking antibodies. Furthermore, ZIKV-mediated fetal pathogenesis was enhanced in mice in the presence of a DENV-reactive monoclonal antibody, but not in the presence of the LALA variant, indicating a dependence on FcγR engagement. Our data suggest a possible mechanism for the recent increase in severe pregnancy outcomes after ZIKV infection in DENV-endemic areas.


Assuntos
Vírus da Dengue/imunologia , Imunidade/imunologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Facilitadores/imunologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Reações Cruzadas/imunologia , Feminino , Humanos , Células K562 , Camundongos , Gravidez , Células Vero
11.
Annu Rev Genet ; 53: 93-116, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31505135

RESUMO

Wolbachia is an endosymbiotic Alphaproteobacteria that can suppress insect-borne diseases through decreasing host virus transmission (population replacement) or through decreasing host population density (population suppression). We contrast natural Wolbachia infections in insect populations with Wolbachia transinfections in mosquitoes to gain insights into factors potentially affecting the long-term success of Wolbachia releases. Natural Wolbachia infections can spread rapidly, whereas the slow spread of transinfections is governed by deleterious effects on host fitness and demographic factors. Cytoplasmic incompatibility (CI) generated by Wolbachia is central to both population replacement and suppression programs, but CI in nature can be variable and evolve, as can Wolbachia fitness effects and virus blocking. Wolbachia spread is also influenced by environmental factors that decrease Wolbachia titer and reduce maternal Wolbachia transmission frequency. More information is needed on the interactions between Wolbachia and host nuclear/mitochondrial genomes, the interaction between invasion success and local ecological factors, and the long-term stability of Wolbachia-mediated virus blocking.


Assuntos
Controle de Doenças Transmissíveis/métodos , Interações Hospedeiro-Patógeno/fisiologia , Insetos Vetores/virologia , Wolbachia/fisiologia , Animais , Evolução Biológica , Citoplasma , Meio Ambiente , Aptidão Genética , Insetos Vetores/microbiologia , Insetos/microbiologia , Insetos/virologia , Mosquitos Vetores/microbiologia , Mosquitos Vetores/virologia
12.
Mol Cell ; 74(4): 801-815.e6, 2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-30952515

RESUMO

Interleukin-1 beta (IL-1ß) is a pleiotropic mediator of inflammation and is produced in response to a wide range of stimuli. During infection, IL-1ß production occurs in parallel with the onset of innate antimicrobial defenses, but the contribution of IL-1ß signaling to cell-intrinsic immunity is not defined. Here, we report that exogenous IL-1ß induces interferon regulatory factor 3 (IRF3) activation in human myeloid, fibroblast, and epithelial cells. IRF3 activation by IL-1ß is dependent upon the DNA-sensing pathway adaptor, stimulator of interferon genes (STING), through the recognition of cytosolic mtDNA by cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS). IL-1ß treatment results in interferon (IFN) production and activation of IFN signaling to direct a potent innate immune response that restricts dengue virus infection. This study identifies a new function for IL-1ß in the onset or enhancement of cell-intrinsic immunity, with important implications for cGAS-STING in integrating inflammatory and microbial cues for host defense.


Assuntos
DNA Mitocondrial/efeitos dos fármacos , Inflamação/genética , Interleucina-1beta/farmacologia , Proteínas de Membrana/genética , Nucleotidiltransferases/genética , GMP Cíclico/genética , DNA Mitocondrial/genética , Dengue/tratamento farmacológico , Dengue/genética , Dengue/virologia , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/genética , Vírus da Dengue/patogenicidade , Interações Hospedeiro-Patógeno/genética , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Inflamação/patologia , Inflamação/virologia , Fator Regulador 3 de Interferon/genética , Interferons/biossíntese , Interleucina-1beta/genética , Células Mieloides/virologia , Transdução de Sinais/efeitos dos fármacos
13.
Trends Biochem Sci ; 47(11): 978-988, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35618579

RESUMO

The antiviral defense directed by the RNAi pathway employs distinct specificity and effector mechanisms compared with other immune responses. The specificity of antiviral RNAi is programmed by siRNAs processed from virus-derived double-stranded RNA by Dicer endonuclease. Argonaute-containing RNA-induced silencing complex loaded with the viral siRNAs acts as the effector to mediate specific virus clearance by RNAi. Recent studies have provided evidence for the production and antiviral function of virus-derived siRNAs in both undifferentiated and differentiated mammalian cells infected with a range of RNA viruses when the cognate virus-encoded suppressor of RNAi (VSR) is rendered nonfunctional. In this review, we discuss the function, mechanism, and evolutionary origin of the validated mammalian VSRs and cell culture assays for their identification.


Assuntos
Proteínas Argonautas , RNA de Cadeia Dupla , Animais , Antivirais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Mamíferos/genética , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Viral/genética
14.
Proc Natl Acad Sci U S A ; 120(41): e2308221120, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37774093

RESUMO

Infants less than 1 y of age experience high rates of dengue disease in dengue virus (DENV) endemic countries. This burden is commonly attributed to antibody-dependent enhancement (ADE), whereby concentrations of maternally derived DENV antibodies become subneutralizing, and infection-enhancing. Understanding antibody-related mechanisms of enhanced infant dengue disease risk represents a significant challenge due to the dynamic nature of antibodies and their imperfect measurement processes. Further, key uncertainties exist regarding the impact of long-term shifts in birth rates, population-level infection risks, and maternal ages on the DENV immune landscape of newborns and their subsequent risks of severe dengue disease in infancy. Here, we analyze DENV antibody data from two infant cohorts (N = 142 infants with 605 blood draws) and 40 y of infant dengue hospitalization data from Thailand. We use mathematical models to reconstruct maternally derived antibody dynamics, accounting for discretized measurement processes and limits of assay detection. We then explore possible antibody-related mechanisms of enhanced infant dengue disease risk and their ability to reconstruct the observed age distribution of hospitalized infant dengue cases. We find that ADE mechanisms are best able to reconstruct the observed data. Finally, we describe how the shifting epidemiology of dengue in Thailand, combined with declining birth rates, have decreased the absolute risk of infant dengue disease by 88% over a 40-y period while having minimal impact on the mean age of infant hospitalized dengue disease.


Assuntos
Vírus da Dengue , Dengue , Dengue Grave , Humanos , Lactente , Recém-Nascido , Anticorpos Antivirais , Anticorpos Neutralizantes , Anticorpos Facilitadores
15.
Proc Natl Acad Sci U S A ; 120(31): e2304667120, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37487061

RESUMO

RNA viruses rapidly adapt to selective conditions due to the high intrinsic mutation rates of their RNA-dependent RNA polymerases (RdRps). Insertions and deletions (indels) in viral genomes are major contributors to both deleterious mutational load and evolutionary novelty, but remain understudied. To characterize the mechanistic details of their formation and evolutionary dynamics during infection, we developed a hybrid experimental-bioinformatic approach. This approach, called MultiMatch, extracts insertions and deletions from ultradeep sequencing experiments, including those occurring at extremely low frequencies, allowing us to map their genomic distribution and quantify the rates at which they occur. Mapping indel mutations in adapting poliovirus and dengue virus populations, we determine the rates of indel generation and identify mechanistic and functional constraints shaping indel diversity. Using poliovirus RdRp variants of distinct fidelity and genome recombination rates, we demonstrate tradeoffs between fidelity and Indel generation. Additionally, we show that maintaining translation frame and viral RNA structures constrain the Indel landscape and that, due to these significant fitness effects, Indels exert a significant deleterious load on adapting viral populations. Conversely, we uncover positively selected Indels that modulate RNA structure, generate protein variants, and produce defective interfering genomes in viral populations. Together, our analyses establish the kinetic and mechanistic tradeoffs between misincorporation, recombination, and Indel rates and reveal functional principles defining the central role of Indels in virus evolution, emergence, and the regulation of viral infection.


Assuntos
Evolução Molecular , Vírus de RNA , Genoma , Taxa de Mutação , Mutação INDEL , RNA Viral/genética , Vírus de RNA/genética
16.
Proc Natl Acad Sci U S A ; 120(3): e2218899120, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36638211

RESUMO

Cleavage of the flavivirus premembrane (prM) structural protein during maturation can be inefficient. The contribution of partially mature flavivirus virions that retain uncleaved prM to pathogenesis during primary infection is unknown. To investigate this question, we characterized the functional properties of newly-generated dengue virus (DENV) prM-reactive monoclonal antibodies (mAbs) in vitro and using a mouse model of DENV disease. Anti-prM mAbs neutralized DENV infection in a virion maturation state-dependent manner. Alanine scanning mutagenesis and cryoelectron microscopy of anti-prM mAbs in complex with immature DENV defined two modes of attachment to a single antigenic site. In vivo, passive transfer of intact anti-prM mAbs resulted in an antibody-dependent enhancement of disease. However, protection against DENV-induced lethality was observed when the transferred mAbs were genetically modified to inhibit their ability to interact with Fcγ receptors. These data establish that in addition to mature forms of the virus, partially mature infectious prM+ virions can also contribute to pathogenesis during primary DENV infections.


Assuntos
Anticorpos Monoclonais , Anticorpos Antivirais , Vírus da Dengue , Dengue , Microscopia Crioeletrônica , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Animais , Camundongos
17.
Proc Natl Acad Sci U S A ; 120(23): e2220005120, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37252973

RESUMO

Dengue virus (DENV) is the most important human virus transmitted by mosquitos. Dengue pathogenesis is characterized by a large induction of proinflammatory cytokines. This cytokine induction varies among the four DENV serotypes (DENV1 to 4) and poses a challenge for live DENV vaccine design. Here, we identify a viral mechanism to limit NF-κB activation and cytokine secretion by the DENV protein NS5. Using proteomics, we found that NS5 binds and degrades the host protein ERC1 to antagonize NF-κB activation, limit proinflammatory cytokine secretion, and reduce cell migration. We found that ERC1 degradation involves unique properties of the methyltransferase domain of NS5 that are not conserved among the four DENV serotypes. By obtaining chimeric DENV2 and DENV4 viruses, we map the residues in NS5 for ERC1 degradation, and generate recombinant DENVs exchanging serotype properties by single amino acid substitutions. This work uncovers a function of the viral protein NS5 to limit cytokine production, critical to dengue pathogenesis. Importantly, the information provided about the serotype-specific mechanism for counteracting the antiviral response can be applied to improve live attenuated vaccines.


Assuntos
Vírus da Dengue , Dengue , Proteínas não Estruturais Virais , Humanos , Citocinas , NF-kappa B/metabolismo , Sorogrupo , Proteínas não Estruturais Virais/metabolismo
18.
Proc Natl Acad Sci U S A ; 120(1): e2211425120, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36577062

RESUMO

De novo viral RNA-dependent RNA polymerases (RdRPs) utilize their priming element (PE) to facilitate accurate initiation. Upon transition to elongation, the PE has to retreat from the active site to give room to the template-product RNA duplex. However, PE conformational change upon this transition and the role of PE at elongation both remain elusive. Here, we report crystal structures of RdRP elongation complex (EC) from dengue virus serotype 2 (DENV2), demonstrating a dramatic refolding of PE that allows establishment of interactions with the RNA duplex backbone approved to be essential for EC stability. Enzymology data from both DENV2 and hepatitis C virus (HCV) RdRPs suggest that critical transition of the refolding likely occurs after synthesis of a 4- to 5-nucleotide (nt) product together providing a key basis in understanding viral RdRP transition from initiation to elongation.


Assuntos
RNA Polimerase Dependente de RNA , RNA , RNA Polimerase Dependente de RNA/metabolismo , Hepacivirus/metabolismo , Domínio Catalítico , Nucleotídeos , RNA Viral/genética
19.
Proc Natl Acad Sci U S A ; 120(42): e2304139120, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37831739

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections are causing significant morbidity and mortality worldwide. Furthermore, over 1 million cases of newly emerging or re-emerging viral infections, specifically dengue virus (DENV), are known to occur annually. Because no virus-specific and fully effective treatments against these or many other viruses have been approved, there is an urgent need for novel, effective therapeutic agents. Here, we identified 2-thiouridine (s2U) as a broad-spectrum antiviral ribonucleoside analogue that exhibited antiviral activity against several positive-sense single-stranded RNA (ssRNA+) viruses, such as DENV, SARS-CoV-2, and its variants of concern, including the currently circulating Omicron subvariants. s2U inhibits RNA synthesis catalyzed by viral RNA-dependent RNA polymerase, thereby reducing viral RNA replication, which improved the survival rate of mice infected with DENV2 or SARS-CoV-2 in our animal models. Our findings demonstrate that s2U is a potential broad-spectrum antiviral agent not only against DENV and SARS-CoV-2 but other ssRNA+ viruses.


Assuntos
Nucleosídeos , Vírus de RNA de Cadeia Positiva , Animais , Camundongos , Nucleosídeos/farmacologia , Antivirais/farmacologia , SARS-CoV-2 , Replicação Viral , RNA
20.
Immunol Rev ; 309(1): 64-74, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35781671

RESUMO

In this review, we discuss how IgG antibodies can modulate inflammatory signaling during viral infections with a focus on CD16a-mediated functions. We describe the structural heterogeneity of IgG antibody ligands, including subclass and glycosylation that impact binding by and downstream activity of CD16a, as well as the heterogeneity of CD16a itself, including allele and expression density. While inflammation is a mechanism required for immune homeostasis and resolution of acute infections, we focus here on two infectious diseases that are driven by pathogenic inflammatory responses during infection. Specifically, we review and discuss the evolving body of literature showing that afucosylated IgG immune complex signaling through CD16a contributes to the overwhelming inflammatory response that is central to the pathogenesis of severe forms of dengue disease and coronavirus disease 2019 (COVID-19).


Assuntos
COVID-19 , Doenças Transmissíveis , Humanos , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Polissacarídeos/química , Polissacarídeos/metabolismo , Receptores de IgG
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA