Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Cell ; 183(4): 1043-1057.e15, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-32970989

RESUMEN

We show that SARS-CoV-2 spike protein interacts with both cellular heparan sulfate and angiotensin-converting enzyme 2 (ACE2) through its receptor-binding domain (RBD). Docking studies suggest a heparin/heparan sulfate-binding site adjacent to the ACE2-binding site. Both ACE2 and heparin can bind independently to spike protein in vitro, and a ternary complex can be generated using heparin as a scaffold. Electron micrographs of spike protein suggests that heparin enhances the open conformation of the RBD that binds ACE2. On cells, spike protein binding depends on both heparan sulfate and ACE2. Unfractionated heparin, non-anticoagulant heparin, heparin lyases, and lung heparan sulfate potently block spike protein binding and/or infection by pseudotyped virus and authentic SARS-CoV-2 virus. We suggest a model in which viral attachment and infection involves heparan sulfate-dependent enhancement of binding to ACE2. Manipulation of heparan sulfate or inhibition of viral adhesion by exogenous heparin presents new therapeutic opportunities.


Asunto(s)
Betacoronavirus/fisiología , Heparitina Sulfato/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Secuencia de Aminoácidos , Enzima Convertidora de Angiotensina 2 , Betacoronavirus/aislamiento & purificación , Sitios de Unión , COVID-19 , Línea Celular , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Heparina/química , Heparina/metabolismo , Heparitina Sulfato/química , Humanos , Riñón/metabolismo , Pulmón/metabolismo , Simulación de Dinámica Molecular , Pandemias , Peptidil-Dipeptidasa A/química , Neumonía Viral/patología , Neumonía Viral/virología , Unión Proteica , Dominios Proteicos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Internalización del Virus
2.
Nature ; 583(7814): E15, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32541969

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
PLoS Pathog ; 18(10): e1010892, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36191050

RESUMEN

Many viruses encode ion channel proteins that oligomerize to form hydrophilic pores in membranes of virus-infected cells and the viral membrane in some enveloped viruses. Alphavirus 6K, human immunodeficiency virus type 1 Vpu (HIV-Vpu), influenza A virus M2 (IAV-M2), and hepatitis C virus P7 (HCV-P7) are transmembrane ion channel proteins that play essential roles in virus assembly, budding, and entry. While the oligomeric structures and mechanisms of ion channel activity are well-established for M2 and P7, these remain unknown for 6K. Here we investigated the functional role of the ion channel activity of 6K in alphavirus assembly by utilizing a series of Sindbis virus (SINV) ion channel chimeras expressing the ion channel helix from Vpu or M2 or substituting the entire 6K protein with full-length P7, in cis. We demonstrate that the Vpu helix efficiently complements 6K, whereas M2 and P7 are less efficient. Our results indicate that while SINV is primarily insensitive to the M2 ion channel inhibitor amantadine, the Vpu inhibitor 5-N, N-Hexamethylene amiloride (HMA), significantly reduces SINV release, suggesting that the ion channel activity of 6K similar to Vpu, promotes virus budding. Using live-cell imaging of SINV with a miniSOG-tagged 6K and mCherry-tagged E2, we further demonstrate that 6K and E2 colocalize with the Golgi apparatus in the secretory pathway. To contextualize the localization of 6K in the Golgi, we analyzed cells infected with SINV and SINV-ion channel chimeras using transmission electron microscopy. Our results provide evidence for the first time for the functional role of 6K in type II cytopathic vacuoles (CPV-II) formation. We demonstrate that in the absence of 6K, CPV-II, which originates from the Golgi apparatus, is not detected in infected cells, with a concomitant reduction in the glycoprotein transport to the plasma membrane. Substituting a functional ion channel, M2 or Vpu localizing to Golgi, restores CPV-II production, whereas P7, retained in the ER, is inadequate to induce CPV-II formation. Altogether our results indicate that ion channel activity of 6K is required for the formation of CPV-II from the Golgi apparatus, promoting glycoprotein spike transport to the plasma membrane and efficient virus budding.


Asunto(s)
Virus Sindbis , Liberación del Virus , Amantadina/farmacología , Glicoproteínas/metabolismo , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Virus Sindbis/genética , Virus Sindbis/metabolismo
4.
Nature ; 562(7725): E3, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29980769

RESUMEN

Change history: In the HTML version of this Letter, Extended Data Fig. 4 incorrectly corresponded to Fig. 4 (the PDF version of the figure was correct). This has been corrected online.

5.
Nature ; 558(7711): 610-614, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29925952

RESUMEN

Viral infections continue to represent major challenges to public health, and an enhanced mechanistic understanding of the processes that contribute to viral life cycles is necessary for the development of new therapeutic strategies 1 . Viperin, a member of the radical S-adenosyl-L-methionine (SAM) superfamily of enzymes, is an interferon-inducible protein implicated in the inhibition of replication of a broad range of RNA and DNA viruses, including dengue virus, West Nile virus, hepatitis C virus, influenza A virus, rabies virus 2 and HIV3,4. Viperin has been suggested to elicit these broad antiviral activities through interactions with a large number of functionally unrelated host and viral proteins3,4. Here we demonstrate that viperin catalyses the conversion of cytidine triphosphate (CTP) to 3'-deoxy-3',4'-didehydro-CTP (ddhCTP), a previously undescribed biologically relevant molecule, via a SAM-dependent radical mechanism. We show that mammalian cells expressing viperin and macrophages stimulated with IFNα produce substantial quantities of ddhCTP. We also establish that ddhCTP acts as a chain terminator for the RNA-dependent RNA polymerases from multiple members of the Flavivirus genus, and show that ddhCTP directly inhibits replication of Zika virus in vivo. These findings suggest a partially unifying mechanism for the broad antiviral effects of viperin that is based on the intrinsic enzymatic properties of the protein and involves the generation of a naturally occurring replication-chain terminator encoded by mammalian genomes.


Asunto(s)
Antivirales/metabolismo , Citidina Trifosfato/metabolismo , Genoma Humano/genética , Proteínas/genética , Proteínas/metabolismo , Terminación de la Transcripción Genética , Animales , Antivirales/química , Chlorocebus aethiops , Citidina Trifosfato/biosíntesis , Citidina Trifosfato/química , Células HEK293 , Humanos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/metabolismo , Ribonucleótidos , Especificidad por Sustrato , Células Vero , Virus Zika/enzimología , Virus Zika/metabolismo
6.
Biochem Soc Trans ; 50(1): 151-165, 2022 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-35015073

RESUMEN

SARS-CoV-2, the coronavirus responsible for the current COVID-19 pandemic, encodes two proteases, 3CLpro and PLpro, two of the main antiviral research targets. Here we provide an overview of the structures and functions of 3CLpro and PLpro and examine strategies of structure-based drug designing and drug repurposing against these proteases. Rational structure-based drug design enables the generation of potent and target-specific antivirals. Drug repurposing offers an attractive prospect with an accelerated turnaround. Thus far, several protease inhibitors have been identified, and some candidates are undergoing trials that may well prove to be effective antivirals against SARS-CoV-2.


Asunto(s)
Reposicionamiento de Medicamentos , Inhibidores de Proteasas/farmacología , SARS-CoV-2/enzimología , Proteínas Virales/antagonistas & inhibidores , Antivirales/farmacología , Diseño de Fármacos , Humanos , Péptido Hidrolasas
7.
Bioessays ; 37(5): 489-94, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25761098

RESUMEN

We highlight the various domains of the flavivirus virulence factor NS1 and speculate on potential implications of the NS1 3D structure in understanding its role in flavivirus pathogenesis. Flavivirus non-structural protein 1 (NS1) is a virulence factor with dual functions in genome replication and immune evasion. Crystal structures of NS1, combined with reconstructions from electron microscopy (EM), provide insight into the architecture of dimeric NS1 on cell membranes and the assembly of a secreted hexameric NS1-lipid complex found in patient sera. Three structural domains of NS1 likely have distinct roles in membrane association, replication complex assembly, and immune system avoidance. A conserved hydrophobic inner face is sequestered either on the membrane or in the interior of the secreted hexamer and contains regions implicated in viral replication. The exposed variable outer face is presented to cellular and secreted components of the immune system in infected patients and contains candidate regions for immune system modulation. We anticipate that knowledge of the distinct NS1 domains and assembly will lead to advances in elucidating virus-host interactions mediated through NS1 and in dissecting the role of NS1 in viral genome replication.


Asunto(s)
Infecciones por Flavivirus/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Microscopía Electrónica de Transmisión , Proteínas no Estructurales Virales/ultraestructura , Replicación Viral/fisiología
8.
Nature ; 468(7324): 705-8, 2010 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-21124457

RESUMEN

Alphaviruses are enveloped RNA viruses that have a diameter of about 700 Å and can be lethal human pathogens. Entry of virus into host cells by endocytosis is controlled by two envelope glycoproteins, E1 and E2. The E2-E1 heterodimers form 80 trimeric spikes on the icosahedral virus surface, 60 with quasi-three-fold symmetry and 20 coincident with the icosahedral three-fold axes arranged with T = 4 quasi-symmetry. The E1 glycoprotein has a hydrophobic fusion loop at one end and is responsible for membrane fusion. The E2 protein is responsible for receptor binding and protects the fusion loop at neutral pH. The lower pH in the endosome induces the virions to undergo an irreversible conformational change in which E2 and E1 dissociate and E1 forms homotrimers, triggering fusion of the viral membrane with the endosomal membrane and then releasing the viral genome into the cytoplasm. Here we report the structure of an alphavirus spike, crystallized at low pH, representing an intermediate in the fusion process and clarifying the maturation process. The trimer of E2-E1 in the crystal structure is similar to the spikes in the neutral pH virus except that the E2 middle region is disordered, exposing the fusion loop. The amino- and carboxy-terminal domains of E2 each form immunoglobulin-like folds, consistent with the receptor attachment properties of E2.


Asunto(s)
Virus Sindbis/química , Virus Sindbis/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Animales , Línea Celular , Microscopía por Crioelectrón , Cristalografía por Rayos X , Drosophila melanogaster , Endosomas/metabolismo , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Fusión de Membrana , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Receptores Virales/metabolismo , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo , Virión/química , Virión/metabolismo
9.
J Virol ; 88(17): 9616-23, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24920796

RESUMEN

UNLABELLED: Alphaviruses are serious, sometimes lethal human pathogens that belong to the family Togaviridae. The structures of human Venezuelan equine encephalitis virus (VEEV), an alphavirus, in complex with two strongly neutralizing antibody Fab fragments (F5 and 3B4C-4) have been determined using a combination of cryo-electron microscopy and homology modeling. We characterize these monoclonal antibody Fab fragments, which are known to abrogate VEEV infectivity by binding to the E2 (envelope) surface glycoprotein. Both of these antibody Fab fragments cross-link the surface E2 glycoproteins and therefore probably inhibit infectivity by blocking the conformational changes that are required for making the virus fusogenic. The F5 Fab fragment cross-links E2 proteins within one trimeric spike, whereas the 3B4C-4 Fab fragment cross-links E2 proteins from neighboring spikes. Furthermore, F5 probably blocks the receptor-binding site, whereas 3B4C-4 sterically hinders the exposure of the fusion loop at the end of the E2 B-domain. IMPORTANCE: Alphaviral infections are transmitted mainly by mosquitoes. Venezuelan equine encephalitis virus (VEEV) is an alphavirus with a wide distribution across the globe. No effective vaccines exist for alphaviral infections. Therefore, a better understanding of VEEV and its associated neutralizing antibodies will help with the development of effective drugs and vaccines.


Asunto(s)
Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/química , Virus de la Encefalitis Equina Venezolana/química , Sustancias Macromoleculares/química , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Simulación por Computador , Microscopía por Crioelectrón , Virus de la Encefalitis Equina Venezolana/inmunología , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/inmunología , Modelos Moleculares , Unión Proteica , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología
10.
J Virol ; 87(15): 8511-23, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23720714

RESUMEN

Alphavirus dogma has long dictated the production of a discrete set of structural proteins during infection of a cell: capsid, pE2, 6K, and E1. However, bioinformatic analyses of alphavirus genomes (A. E. Firth, B. Y. Chung, M. N. Fleeton, and J. F. Atkins, Virol. J. 5:108, 2008) suggested that a ribosomal frameshifting event occurs during translation of the alphavirus structural polyprotein. Specifically, a frameshift event is suggested to occur during translation of the 6K gene, yielding production of a novel protein, termed transframe (TF), comprised of a C-terminal extension of the 6K protein in the -1 open reading frame (ORF). Here, we validate the findings of Firth and colleagues with respect to the production of the TF protein and begin to characterize the function of TF. Using a mass spectrometry-based approach, we identified TF in purified preparations of both Sindbis and Chikungunya virus particles. We next constructed a panel of Sindbis virus mutants with mutations which alter the production, size, or sequence of TF. We demonstrate that TF is not absolutely required in culture, although disrupting TF production leads to a decrease in virus particle release in both mammalian and insect cells. In a mouse neuropathogenesis model, mortality was <15% in animals infected with the TF mutants, whereas mortality was 95% in animals infected with the wild-type virus. Using a variety of additional assays, we demonstrate that TF retains ion-channel activity analogous to that of 6K and that lack of production of TF does not affect genome replication, particle infectivity, or envelope protein transit to the cell surface. The TF protein therefore represents a previously uncharacterized factor important for alphavirus assembly.


Asunto(s)
Virus Chikungunya/fisiología , Regulación Viral de la Expresión Génica , Virus Sindbis/fisiología , Proteínas Virales/biosíntesis , Ensamble de Virus , Infecciones por Alphavirus/mortalidad , Infecciones por Alphavirus/patología , Infecciones por Alphavirus/virología , Animales , Línea Celular , Virus Chikungunya/genética , Virus Chikungunya/patogenicidad , Modelos Animales de Enfermedad , Insectos , Ratones , Virus Sindbis/genética , Análisis de Supervivencia , Replicación Viral
11.
Pathogens ; 13(2)2024 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-38392858

RESUMEN

The Flavivirus genus of the Flaviviridae family of enveloped single-stranded RNA viruses encompasses more than 70 members, many of which cause significant disease in humans and livestock. Packaging and assembly of the flavivirus RNA genome is essential for the formation of virions, which requires intricate coordination of genomic RNA, viral structural, and nonstructural proteins in association with virus-induced, modified endoplasmic reticulum (ER) membrane structures. The capsid (C) protein, a small but versatile RNA-binding protein, and the positive single-stranded RNA genome are at the heart of the elusive flavivirus assembly process. The nucleocapsid core, consisting of the genomic RNA encapsidated by C proteins, buds through the ER membrane, which contains viral glycoproteins prM and E organized as trimeric spikes into the lumen, forming an immature virus. During the maturation process, which involves the low pH-mediated structural rearrangement of prM and E and furin cleavage of prM in the secretory pathway, the spiky immature virus with a partially ordered nucleocapsid core becomes a smooth, mature virus with no discernible nucleocapsid. This review focuses on the mechanisms of genome packaging and assembly by examining the structural and functional aspects of C protein and viral RNA. We review the current lexicon of critical C protein features and evaluate interactions between C and genomic RNA in the context of assembly and throughout the life cycle.

12.
PLoS Negl Trop Dis ; 18(1): e0011873, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38166143

RESUMEN

Flaviviruses such as Zika and dengue viruses are persistent health concerns in endemic regions worldwide. Efforts to combat the spread of flaviviruses have been challenging, as no antivirals or optimal vaccines are available. Prevention and treatment of flavivirus-induced diseases require a comprehensive understanding of their life cycle. However, several aspects of flavivirus biogenesis, including genome packaging and virion assembly, are not well characterized. In this study, we focused on flavivirus capsid protein (C) using Zika virus (ZIKV) as a model to investigate the role of the externally oriented α3 helix (C α3) without a known or predicted function. Alanine scanning mutagenesis of surface-exposed amino acids on C α3 revealed a critical CN67 residue essential for ZIKV virion production. The CN67A mutation did not affect dimerization or RNA binding of purified C protein in vitro. The virus assembly is severely affected in cells transfected with an infectious cDNA clone of ZIKV with CN67A mutation, resulting in a highly attenuated phenotype. We isolated a revertant virus with a partially restored phenotype by continuous passage of the CN67A mutant virus in Vero E6 cells. Sequence analysis of the revertant revealed a second site mutation in the viral membrane (M) protein MF37L, indicating a genetic interaction between the C and M proteins of ZIKV. Introducing the MF37L mutation on the mutant ZIKV CN67A generated a double-mutant virus phenotypically consistent with the isolated genetic revertant. Similar results were obtained with analogous mutations on C and M proteins of dengue virus, suggesting the critical nature of C α3 and possible C and M residues contributing to virus assembly in other Aedes-transmitted flaviviruses. This study provides the first experimental evidence of a genetic interaction between the C protein and the viral envelope protein M, providing a mechanistic understanding of the molecular interactions involved in the assembly and budding of Aedes-transmitted flaviviruses.


Asunto(s)
Aedes , Flavivirus , Infección por el Virus Zika , Virus Zika , Animales , Cápside , Proteínas de la Cápside/genética , Ensamble de Virus/genética , Replicación Viral/genética , Virus Zika/genética
13.
J Virol ; 86(5): 2585-99, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22190727

RESUMEN

Alphavirus budding from the plasma membrane occurs through the specific interaction of the nucleocapsid core with the cytoplasmic domain of the E2 glycoprotein (cdE2). Structural studies of the Sindbis virus capsid protein (CP) have suggested that these critical interactions are mediated by the binding of cdE2 into a hydrophobic pocket in the CP. Several molecular genetic studies have implicated amino acids Y400 and L402 in cdE2 as important for the budding of alphaviruses. In this study, we characterized the role of cdE2 residues in structural polyprotein processing, glycoprotein transport, and capsid interactions. Along with hydrophobic residues, charged residues in the N terminus of cdE2 were critical for the effective interaction of cores with cdE2, a process required for virus budding. Mutations in the C-terminal signal sequence region of cdE2 affected E2 protein transport to the plasma membrane, while nonbudding mutants that were defective in cdE2-CP interaction accumulated E2 on the plasma membrane. The interaction of cdE2 with cytoplasmic cores purified from infected cells and in vitro-assembled core-like particles suggests that cdE2 interacts with assembled cores to mediate budding. We hypothesize that these cdE2 interactions induce a change in the organization of the nucleocapsid core upon binding leading to particle budding and priming of the nucleocapsid cores for disassembly that is required for virus infection.


Asunto(s)
Infecciones por Alphavirus/virología , Proteínas de la Cápside/metabolismo , Virus Sindbis/fisiología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Liberación del Virus , Proteínas de la Cápside/genética , Línea Celular , Humanos , Unión Proteica , Estructura Terciaria de Proteína , Virus Sindbis/química , Virus Sindbis/genética , Proteínas del Envoltorio Viral/genética
14.
J Virol ; 86(22): 12372-83, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22951842

RESUMEN

A 7-Å cryoelectron microscopy-based reconstruction of Sindbis virus (SINV) was recently generated. Fitting the crystal structure of the SINV capsid protein (Cp) into the density map revealed that the F2-G2 loop of the Cp was shifted away from cytoplasmic domain of E2 (cdE2) in the 7-Å reconstruction relative to its position in the Cp crystal structure. Furthermore, the reconstruction demonstrated that residue E395 in region I of the cytoplasmic domain of the E2 envelope protein (cdE2-RI) and K252 of Cp, part of the Cp F2-G2 loop, formed a putative salt bridge in the virion. We generated amino acid substitutions at residues K250 and K252 of the SINV Cp and explored the resulting phenotypes. In the context of cells infected with wild-type or mutant virus, reversing the charge of these two residues resulted in the appearance of Cp aggregates around cytopathic vacuole type I (CPV-I) structures, the absence of nucleocapsid (NC) formation, and a lack of virus particle release in the infected mammalian cell. However, expressing the same Cp mutants in the cell without the envelope proteins or expressing and purifying the mutants from an Escherichia coli expression system and assembling in vitro yielded NC assembly in all cases. In addition, second-site mutations within cdE2 restored NC assembly but not release of infectious particles. Our data suggest an early temporal and spatial interaction between cdE2-RI and the Cp F2-G2 loop that, when ablated, leads to the absence of NC assembly. This interaction also appears to be important for budding of virus particles.


Asunto(s)
Cápside/química , Virus Sindbis/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Secuencias de Aminoácidos , Animales , Cápside/metabolismo , Línea Celular , Cricetinae , Microscopía por Crioelectrón/métodos , Cristalografía por Rayos X/métodos , Dimerización , Ensayo de Inmunoadsorción Enzimática/métodos , Microscopía Electrónica/métodos , Mutación , Fenotipo , Estructura Terciaria de Proteína
15.
Zootaxa ; 5258(3): 270-284, 2023 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-37044595

RESUMEN

A new genus and species, Capulopsyche keralensis gen. et sp. nov. from the subfamily Taleporiinae is described from Kerala, India. The new genus has distinct features from other genera of the subfamily. Double-walled larval case architecture is one of the unique characteristics of this genus. Details of life stages, distribution, COI sequencing, and the morphology of adults are described in the paper.


Asunto(s)
Lepidópteros , Animales , Larva , India
16.
Zootaxa ; 5352(4): 521-536, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-38221431

RESUMEN

A new species of bagworm moth, Eumasia venefica sp. nov., from the subfamily Eumasiinae, is described from Kerala, India. This species has a peculiar larval ecology, larval case structure and an association with lichens. The paper describes the detailed taxonomy of the adult and the larval stages of the species.


Asunto(s)
Lepidópteros , Mariposas Nocturnas , Animales , Larva , India
17.
Res Sq ; 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-38106210

RESUMEN

Zika virus (ZIKV) infection continues to pose a significant public health concern due to limited available preventive measures and treatments. ZIKV is unique among flaviviruses in its vertical transmission capacity (i.e., transmission from mother to fetus) yet the underlying mechanisms remain incompletely understood. Here, we show that both African and Asian lineages of ZIKV induce tunneling nanotubes (TNTs) in placental trophoblasts and multiple other mammalian cell types. Amongst investigated flaviviruses, only ZIKV strains trigger TNTs. We show that ZIKV-induced TNTs facilitate transfer of viral particles, proteins, and RNA to neighboring uninfected cells. ZIKV TNT formation is driven exclusively via its non-structural protein 1 (NS1); specifically, the N-terminal region (50 aa) of membrane-bound NS1 is necessary and sufficient for triggering TNT formation in host cells. Using affinity purification-mass spectrometry of cells infected with wild-type NS1 or non-TNT forming NS1 (pNS1ΔTNT) proteins, we found mitochondrial proteins are dominant NS1-interacting partners, consistent with the elevated mitochondrial mass we observed in infected trophoblasts. We demonstrate that mitochondria are siphoned via TNTs from healthy to ZIKV-infected cells, both homotypically and heterotypically, and inhibition of mitochondrial respiration reduced viral replication in trophoblast cells. Finally, ZIKV strains lacking TNT capabilities due to mutant NS1 elicited a robust antiviral IFN-λ 1/2/3 response, indicating ZIKV's TNT-mediated trafficking also allows ZIKV cell-cell transmission that is camouflaged from host defenses. Together, our findings identify a new stealth mechanism that ZIKV employs for intercellular spread among placental trophoblasts, evasion of antiviral interferon response, and the hijacking of mitochondria to augment its propagation and survival. Discerning the mechanisms of ZIKV intercellular strategies offers a basis for novel therapeutic developments targeting these interactions to limit its dissemination.

18.
J Virol ; 85(12): 5773-81, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21471237

RESUMEN

Alphaviruses are small, spherical, enveloped, positive-sense, single-stranded, RNA viruses responsible for considerable human and animal disease. Using microinjection of preassembled cores as a tool, a system has been established to study the assembly and budding process of Sindbis virus, the type member of the alphaviruses. We demonstrate the release of infectious virus-like particles from cells expressing Sindbis virus envelope glycoproteins following microinjection of Sindbis virus nucleocapsids purified from the cytoplasm of infected cells. Furthermore, it is shown that nucleocapsids assembled in vitro mimic those isolated in the cytoplasm of infected cells with respect to their ability to be incorporated into enveloped virions following microinjection. This system allows for the study of the alphavirus budding process independent of an authentic infection and provides a platform to study viral and host requirements for budding.


Asunto(s)
Riñón/virología , Nucleocápside/administración & dosificación , Virión/fisiología , Ensamble de Virus/fisiología , Liberación del Virus/fisiología , Alphavirus/genética , Alphavirus/metabolismo , Alphavirus/fisiología , Animales , Línea Celular , Cricetinae , Humanos , Riñón/citología , Microinyecciones , Nucleocápside/genética , Nucleocápside/aislamiento & purificación , Nucleocápside/metabolismo , Virus Sindbis/metabolismo , Virus Sindbis/fisiología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Virión/patogenicidad , Virología/métodos
19.
Curr Opin Virol ; 54: 101230, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35576774

RESUMEN

Asymmetric structural elements are typically not readily visualized in icosahedral viruses that have other obvious symmetrical features and most asymmetry has gone unresolved for decades. Asymmetric features may be incorporated during assembly or maturation or develop during key steps in the infectious cycle of the virus. However, resolving asymmetric features requires abandoning capsid-wide symmetry averaging and relying on special applications during single-particle cryogenic electron microscopy (cryo-EM) analysis. Thanks to the advances in the cryo-EM field, we are learning more about asymmetry of viruses. Here we summarize some of what is currently known about asymmetric structural features using as examples members of the Togaviridae, Flaviviridae, Herpesviridae, Parvoviridae, and Papillomaviridae.


Asunto(s)
Virión , Virus , Cápside/química , Proteínas de la Cápside/química , Microscopía por Crioelectrón , Virión/química , Virus/química , Virus/genética
20.
Zootaxa ; 5159(1): 136-144, 2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-36095554

RESUMEN

Acanthopsyche alstoni Watt Mann, is a lesser known psychid previously known only from the description of its larval cases collected from Assam, India in 1903. There were no descriptions of the adult moth or biology of the species. During surveys for Psychidae in the human inhabited regions of Kerala State, India, more than 100 larval cases were recovered. They were allowed to hatch and characteristics were recorded. This paper provides a detailed redescription of this species and designation of a neotype.


Asunto(s)
Mariposas Nocturnas , Animales , Biología , Humanos , India , Larva
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA