Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Virus Res ; 345: 199398, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38754786

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne nairovirus with a wide geographic spread that can cause severe and lethal disease. No specific medical countermeasures are approved to combat this illness. The CCHFV L protein contains an ovarian tumor (OTU) domain with a cysteine protease thought to modulate cellular immune responses by removing ubiquitin and ISG15 post-translational modifications from host and viral proteins. Viral deubiquitinases like CCHFV OTU are attractive drug targets, as blocking their activity may enhance cellular immune responses to infection, and potentially inhibit viral replication itself. We previously demonstrated that the engineered ubiquitin variant CC4 is a potent inhibitor of CCHFV replication in vitro. A major challenge of the therapeutic use of small protein inhibitors such as CC4 is their requirement for intracellular delivery, e.g., by viral vectors. In this study, we examined the feasibility of in vivo CC4 delivery by a replication-deficient recombinant adenovirus (Ad-CC4) in a lethal CCHFV mouse model. Since the liver is a primary target of CCHFV infection, we aimed to optimize delivery to this organ by comparing intravenous (tail vein) and intraperitoneal injection of Ad-CC4. While tail vein injection is a traditional route for adenovirus delivery, in our hands intraperitoneal injection resulted in higher and more widespread levels of adenovirus genome in tissues, including, as intended, the liver. However, despite promising in vitro results, neither route of in vivo CC4 treatment resulted in protection from a lethal CCHFV infection.


Assuntos
Adenoviridae , Modelos Animais de Doenças , Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Replicação Viral , Animais , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Febre Hemorrágica da Crimeia/virologia , Camundongos , Adenoviridae/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vetores Genéticos/genética , Antivirais/farmacologia , Feminino , Fígado/virologia , Humanos
2.
Virus Res ; 286: 198036, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32492472

RESUMO

Interferon (IFN)-stimulated gene product 15 (ISG15) is a ubiquitin-like protein critical for the control of microbial infections. ISG15 appears to serve a wide variety of functions, which regulate multiple cellular responses contributing to the development of an antiviral state. ISG15 is a versatile molecule directly modulating both host and virus protein function which regulate many signaling pathways, including its own synthesis. Here we review the various roles ISG15 plays in the antiviral immune response, and examine the mechanisms by which viruses attempt to mitigate or exploit ISG15 activity.


Assuntos
Citocinas/metabolismo , Imunidade Inata/imunologia , Ubiquitinas/metabolismo , Viroses/imunologia , Replicação Viral/imunologia , Animais , Citocinas/genética , Humanos , Interferon Tipo I/imunologia , Macrófagos/imunologia , Camundongos , Neutrófilos/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Ubiquitinas/genética , Proteínas Virais/metabolismo , Internalização do Vírus
3.
PLoS Pathog ; 15(12): e1008183, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31790513

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV, order Bunyavirales, family Nairoviridae, genus Orthonairovirus) is the tick-borne etiological agent of Crimean-Congo hemorrhagic fever (CCHF) in humans. Animals are generally susceptible to CCHFV infection but refractory to disease. Small animal models are limited to interferon-deficient mice, that develop acute fatal disease following infection. Here, using a ZsGreen1- (ZsG) expressing reporter virus (CCHFV/ZsG), we examine tissue tropism and dissemination of virus in interferon-α/ß receptor knock-out (Ifnar-/-) mice. We demonstrate that CCHFV/ZsG retains in vivo pathogenicity comparable to wild-type virus. Interestingly, despite high levels of viral RNA in all organs assessed, 2 distribution patterns of infection were observed by both fluorescence and immunohistochemistry (IHC), corresponding to the permissiveness of organ tissues. To further investigate viral dissemination and to temporally define cellular targets of CCHFV in vivo, mice were serially euthanized at different stages of disease. Flow cytometry was used to characterize CCHFV-associated alterations in hematopoietic cell populations and to classify infected cells in the blood, lymph node, spleen, and liver. ZsG signal indicated that mononuclear phagocytic cells in the lymphatic tissues were early targets of infection; in late-stage infection, overall, the highest levels of signal were detected in the liver, and ZsG was found in both antigen-presenting and lymphocyte cell populations.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia/urina , Sistema Fagocitário Mononuclear/virologia , Tropismo Viral/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência
4.
PLoS One ; 14(12): e0226415, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31869347

RESUMO

Tick-borne nairoviruses (order Bunyavirales) encode an ovarian tumor domain protease (OTU) that suppresses the innate immune response by reversing the post-translational modification of proteins by ubiquitin (Ub) and interferon-stimulated gene product 15 (ISG15). Ub is highly conserved across eukaryotes, whereas ISG15 is only present in vertebrates and shows substantial sequence diversity. Prior attempts to address the effect of ISG15 diversity on viral protein-ISG15 interactions have focused on only a single species' ISG15 or a limited selection of nairovirus OTUs. To gain a more complete perspective of OTU-ISG15 interactions, we biochemically assessed the relative activities of 14 diverse nairovirus OTUs for 12 species' ISG15 and found that ISG15 activity is predominantly restricted to particular nairovirus lineages reflecting, in general, known virus-host associations. To uncover the underlying molecular factors driving OTUs affinity for ISG15, X-ray crystal structures of Kupe virus and Ganjam virus OTUs bound to sheep ISG15 were solved and compared to complexes of Crimean-Congo hemorrhagic fever virus and Erve virus OTUs bound to human and mouse ISG15, respectively. Through mutational and structural analysis seven residues in ISG15 were identified that predominantly influence ISG15 species specificity among nairovirus OTUs. Additionally, OTU residues were identified that influence ISG15 preference, suggesting the potential for viral OTUs to adapt to different host ISG15s. These findings provide a foundation to further develop research methods to trace nairovirus-host relationships and delineate the full impact of ISG15 diversity on nairovirus infection.


Assuntos
Citocinas/metabolismo , Especificidade de Hospedeiro/genética , Nairovirus/enzimologia , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Domínios e Motivos de Interação entre Proteínas/genética , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Animais , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Cristalografia por Raios X , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Humanos , Camundongos , Modelos Moleculares , Nairovirus/classificação , Nairovirus/genética , Peptídeo Hidrolases/química , Filogenia , Ligação Proteica/genética , Homologia de Sequência , Ovinos , Especificidade da Espécie , Ubiquitina/metabolismo , Proteínas Virais/química , Proteínas Virais/genética
5.
mBio ; 10(4)2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31337717

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV) infection can result in a severe hemorrhagic syndrome for which there are no antiviral interventions available to date. Certain RNA viruses, such as CCHFV, encode cysteine proteases of the ovarian tumor (OTU) family that antagonize interferon (IFN) production by deconjugating ubiquitin (Ub). The OTU of CCHFV, a negative-strand RNA virus, is dispensable for replication of the viral genome, despite being part of the large viral RNA polymerase. Here, we show that mutations that prevent binding of the OTU to cellular ubiquitin are required for the generation of recombinant CCHFV containing a mutated catalytic cysteine. Similarly, the high-affinity binding of a synthetic ubiquitin variant (UbV-CC4) to CCHFV OTU strongly inhibits viral growth. UbV-CC4 inhibits CCHFV infection even in the absence of intact IFN signaling, suggesting that its antiviral activity is not due to blocking the OTU's immunosuppressive function. Instead, the prolonged occupancy of the OTU with UbV-CC4 directly targets viral replication by interfering with CCHFV RNA synthesis. Together, our data provide mechanistic details supporting the development of antivirals targeting viral OTUs.IMPORTANCE Crimean-Congo hemorrhagic fever virus is an important human pathogen with a wide global distribution for which no therapeutic interventions are available. CCHFV encodes a cysteine protease belonging to the ovarian tumor (OTU) family which is involved in host immune suppression. Here we demonstrate that artificially prolonged binding of the OTU to a substrate inhibits virus infection. This provides novel insights into CCHFV OTU function during the viral replicative cycle and highlights the OTU as a potential antiviral target.


Assuntos
Cisteína Proteases/metabolismo , Enzimas Desubiquitinantes/metabolismo , Vírus da Febre Hemorrágica da Crimeia-Congo/enzimologia , Ubiquitina/farmacologia , Replicação Viral , Animais , Linhagem Celular Tumoral , Cisteína Proteases/genética , Citocinas/genética , Citocinas/metabolismo , Enzimas Desubiquitinantes/genética , Feminino , Vírus da Febre Hemorrágica da Crimeia-Congo/fisiologia , Humanos , Camundongos , Mutação , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Ubiquitinação , Ubiquitinas/genética , Ubiquitinas/metabolismo
6.
PLoS Pathog ; 15(1): e1007515, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30629698

RESUMO

Post-translational modification of host and viral proteins by ubiquitin (Ub) and Ub-like proteins, such as interferon stimulated gene product 15 (ISG15), plays a key role in response to infection. Viruses have been increasingly identified that contain proteases possessing deubiquitinase (DUB) and/or deISGylase functions. This includes viruses in the Nairoviridae family that encode a viral homologue of the ovarian tumor protease (vOTU). vOTU activity was recently demonstrated to be critical for replication of the often-fatal Crimean-Congo hemorrhagic fever virus, with DUB activity suppressing the type I interferon responses and deISGylase activity broadly removing ISG15 conjugated proteins. There are currently about 40 known nairoviruses classified into fourteen species. Recent genomic characterization has revealed a high degree of diversity, with vOTUs showing less than 25% amino acids identities within the family. Previous investigations have been limited to only a few closely related nairoviruses, leaving it unclear what impact this diversity has on vOTU function. To probe the effects of vOTU diversity on enzyme activity and specificity, we assessed representative vOTUs spanning the Nairoviridae family towards Ub and ISG15 fluorogenic substrates. This revealed great variation in enzymatic activity and specific substrate preferences. A subset of the vOTUs were further assayed against eight biologically relevant di-Ub substrates, uncovering both common trends and distinct preferences of poly-Ub linkages by vOTUs. Four novel X-ray crystal structures were obtained that provide a biochemical rationale for vOTU substrate preferences and elucidate structural features that distinguish the vOTUs, including a motif in the Hughes orthonairovirus species that has not been previously observed in OTU domains. Additionally, structure-informed mutagenesis provided the first direct evidence of a second site involved in di-Ub binding for vOTUs. These results provide new insight into nairovirus evolution and pathogenesis, and further enhances the development of tools for therapeutic purposes.


Assuntos
Nairovirus/genética , Neoplasias Ovarianas/virologia , Peptídeo Hidrolases/genética , Cristalografia por Raios X/métodos , Enzimas Desubiquitinantes/metabolismo , Feminino , Variação Genética/genética , Genômica , Humanos , Nairovirus/patogenicidade , Neoplasias Ovarianas/metabolismo , Ovário/metabolismo , Peptídeo Hidrolases/metabolismo , Filogenia , Ligação Proteica , Domínios Proteicos , Processamento de Proteína Pós-Traducional/genética , Proteólise , Homologia de Sequência de Aminoácidos , Ubiquitina/metabolismo , Ubiquitinação/genética , Ubiquitinas/metabolismo , Proteínas Virais/metabolismo
7.
Antiviral Res ; 147: 91-99, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29024765

RESUMO

Crimean-Congo hemorrhagic fever virus (CCHFV), a tick-borne orthonairovirus, causes a severe hemorrhagic disease in humans (Crimean-Congo hemorrhagic fever, CCHF). Currently, no vaccines are approved to prevent CCHF; treatment is limited to supportive care and the use of ribavirin, the therapeutic benefits of which remain unclear. CCHF is part of WHO's priority list of infectious diseases warranting further research and development. To aid in the identification of new antiviral compounds, we generated a recombinant CCHFV expressing a reporter protein, allowing us to quantify virus inhibition by measuring the reduction in fluorescence in infected cells treated with candidate compounds. The screening assay was readily adaptable to high-throughput screening (HTS) of compounds using Huh7 cells, with a signal-to-noise ratio of 50:1, and Z'-factors > 0.6 in both 96- and 384-well formats. A screen of candidate nucleoside analog compounds identified 2'-deoxy-2'-fluorocytidine (EC50 = 61 ± 18 nM) as having 200 × the potency of ribavirin (EC50 = 12.5 ± 2.6 µM), as well as 17 × the potency of T-705 (favipiravir), another compound with reported anti-CCHFV activity (EC50 = 1.03 ± 0.16 µM). Furthermore, we also determined that 2'-deoxy-2'-fluorocytidine acts synergistically with T-705 to inhibit CCHFV replication without causing cytotoxicity. The incorporation of this reporter virus into the high-throughput screening assay described here will allow more rapid identification of effective therapeutic options to combat this emerging human pathogen.


Assuntos
Desoxicitidina/análogos & derivados , Descoberta de Drogas/métodos , Vírus da Febre Hemorrágica da Crimeia-Congo/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Replicação Viral/efeitos dos fármacos , Amidas/farmacologia , Antivirais/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Genes Reporter/genética , Proteínas de Fluorescência Verde/genética , Febre Hemorrágica da Crimeia/virologia , Humanos , Dose Letal Mediana , Pirazinas/farmacologia , Ribavirina/farmacologia , Proteínas Virais/genética
8.
Cell Rep ; 20(10): 2396-2407, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28877473

RESUMO

Antiviral responses are regulated by conjugation of ubiquitin (Ub) and interferon-stimulated gene 15 (ISG15) to proteins. Certain classes of viruses encode Ub- or ISG15-specific proteases belonging to the ovarian tumor (OTU) superfamily. Their activity is thought to suppress cellular immune responses, but studies demonstrating the function of viral OTU proteases during infection are lacking. Crimean-Congo hemorrhagic fever virus (CCHFV, family Nairoviridae) is a highly pathogenic human virus that encodes an OTU with both deubiquitinase and deISGylase activity as part of the viral RNA polymerase. We investigated CCHFV OTU function by inactivating protease catalytic activity or by selectively disrupting its deubiquitinase and deISGylase activity using reverse genetics. CCHFV OTU inactivation blocked viral replication independently of its RNA polymerase activity, while deubiquitinase activity proved critical for suppressing the interferon responses. Our findings provide insights into viral OTU functions and support the development of therapeutics and vaccines.


Assuntos
Citocinas/metabolismo , Enzimas Desubiquitinantes/metabolismo , Vírus da Febre Hemorrágica da Crimeia-Congo/metabolismo , Neoplasias Ovarianas/imunologia , Proteases Específicas de Ubiquitina/metabolismo , Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Citocinas/genética , Enzimas Desubiquitinantes/genética , Feminino , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Humanos , Neoplasias Ovarianas/metabolismo , Proteases Específicas de Ubiquitina/genética , Ubiquitinas/genética , Replicação Viral/genética , Replicação Viral/fisiologia
9.
J Virol ; 89(8): 4457-69, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25653451

RESUMO

UNLABELLED: Stress granules (SGs) are protein-mRNA aggregates that are formed in response to environmental stresses, resulting in translational inhibition. SGs are generally believed to play an antiviral role and are manipulated by many viruses, including various alphaviruses. GTPase-activating protein (SH3 domain)-binding protein 1 (G3BP1) is a key component and commonly used marker of SGs. Its homolog G3BP2 is a less extensively studied SG component. Here, we demonstrate that Chikungunya virus (CHIKV) infection induces cytoplasmic G3BP1- and G3BP2-containing granules that differ from bona fide SGs in terms of morphology, composition, and behavior. For several Old World alphaviruses it has been shown that nonstructural protein 3 (nsP3) interacts with G3BPs, presumably to inhibit SG formation, and we have confirmed this interaction in CHIKV-infected cells. Surprisingly, CHIKV also relied on G3BPs for efficient replication, as simultaneous depletion of G3BP1 and G3BP2 reduced viral RNA levels, CHIKV protein expression, and viral progeny titers. The G3BPs colocalized with CHIKV nsP2 and nsP3 in cytoplasmic foci, but no colocalization with nsP1, nsP4, or dsRNA was observed. Furthermore, G3BPs could not be detected in a cellular fraction enriched for CHIKV replication/transcription complexes, suggesting that they are not directly involved in CHIKV RNA synthesis. Depletion of G3BPs did not affect viral entry, translation of incoming genomes, or nonstructural polyprotein processing but resulted in severely reduced levels of negative-stranded (and consequently also positive-stranded) RNA. This suggests a role for the G3BPs in the switch from translation to genome amplification, although the exact mechanism by which they act remains to be explored. IMPORTANCE: Chikungunya virus (CHIKV) causes a severe polyarthritis that has affected millions of people since its reemergence in 2004. The lack of approved vaccines or therapeutic options and the ongoing explosive outbreak in the Caribbean underline the importance of better understanding CHIKV replication. Stress granules (SGs) are cytoplasmic protein-mRNA aggregates formed in response to various stresses, including viral infection. The RNA-binding proteins G3BP1 and G3BP2 are essential SG components. SG formation and the resulting translational inhibition are generally considered an antiviral response, and many viruses manipulate or block this process. Late in infection, we and others have observed CHIKV nonstructural protein 3 in cytoplasmic G3BP1- and G3BP2-containing granules. These virally induced foci differed from true SGs and did not appear to represent replication complexes. Surprisingly, we found that G3BP1 and G3BP2 were also needed for efficient CHIKV replication, likely by facilitating the switch from translation to genome amplification early in infection.


Assuntos
Proteínas de Transporte/metabolismo , Febre de Chikungunya/metabolismo , Vírus Chikungunya/fisiologia , Regulação Viral da Expressão Gênica/fisiologia , Replicação Viral/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Western Blotting , Fracionamento Celular , Chlorocebus aethiops , Clonagem Molecular , DNA Helicases , Primers do DNA/genética , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Luciferases , Proteínas de Ligação a Poli-ADP-Ribose , RNA Helicases , Interferência de RNA , Proteínas com Motivo de Reconhecimento de RNA , Proteínas de Ligação a RNA , Reação em Cadeia da Polimerase em Tempo Real , Células Vero , Ensaio de Placa Viral
10.
J Virol ; 88(8): 4180-94, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24478443

RESUMO

UNLABELLED: RIG-I is a cytosolic sensor critically involved in the activation of the innate immune response to RNA virus infection. In the present study, we evaluated the inhibitory effect of a RIG-I agonist on the replication of two emerging arthropod-borne viral pathogens, dengue virus (DENV) and chikungunya virus (CHIKV), for which no therapeutic options currently exist. We demonstrate that when a low, noncytotoxic dose of an optimized 5'triphosphorylated RNA (5'pppRNA) molecule was administered, RIG-I stimulation generated a robust antiviral response against these two viruses. Strikingly, 5'pppRNA treatment before or after challenge with DENV or CHIKV provided protection against infection. In primary human monocytes and monocyte-derived dendritic cells, the RIG-I agonist blocked both primary infection and antibody-dependent enhancement of DENV infection. The protective response against DENV and CHIKV induced by 5'pppRNA was dependent on an intact RIG-I/MAVS/TBK1/IRF3 axis and was largely independent of the type I IFN response. Altogether, this in vitro analysis of the antiviral efficacy of 5'pppRNA highlights the therapeutic potential of RIG-I agonists against emerging viruses such as DENV and CHIKV. IMPORTANCE: DENV and CHIKV are two reemerging mosquito-borne viruses for which no therapeutic options currently exist. Both viruses overlap geographically in tropical regions of the world, produce similar fever-like symptoms, and are difficult to diagnose. This study investigated the inhibitory effect of a RIG-I agonist on the replication of these two viruses. RIG-I stimulation using 5'pppRNA before or after DENV or CHIKV infection generated a protective antiviral response against both pathogens in immune and nonimmune cells; interestingly, the protective response against the viruses was largely independent of the classical type I interferon response. The antiviral efficacy of 5'pppRNA highlights the therapeutic potential of RIG-I agonists against emerging viruses such as DENV and CHIKV.


Assuntos
Infecções por Alphavirus/imunologia , Vírus Chikungunya/fisiologia , RNA Helicases DEAD-box/imunologia , Dengue/imunologia , Imunidade Inata , Interferon Tipo I/imunologia , Infecções por Alphavirus/genética , Infecções por Alphavirus/virologia , Animais , Linhagem Celular , Febre de Chikungunya , Vírus Chikungunya/genética , Vírus Chikungunya/imunologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Dengue/genética , Dengue/virologia , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Vírus da Dengue/fisiologia , Humanos , Interferon Tipo I/genética , Camundongos , Receptores Imunológicos , Replicação Viral
11.
PLoS One ; 8(8): e71047, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23936484

RESUMO

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that re-emerged in 2004 and has caused massive outbreaks in recent years. The lack of a licensed vaccine or treatment options emphasize the need to obtain more insight into the viral life cycle and CHIKV-host interactions. Infectious cDNA clones are important tools for such studies, and for mechanism of action studies on antiviral compounds. Existing CHIKV cDNA clones are based on a single genome from an individual clinical isolate, which is expected to have evolved specific characteristics in response to the host environment, and possibly also during subsequent cell culture passaging. To obtain a virus expected to have the general characteristics of the recent E1-226V CHIKV isolates, we have constructed a new CHIKV full-length cDNA clone, CHIKV LS3, based on the consensus sequence of their aligned genomes. Here we report the characterization of this synthetic virus and a green fluorescent protein-expressing variant (CHIKV LS3-GFP). Their characteristics were compared to those of natural strain ITA07-RA1, which was isolated during the 2007 outbreak in Italy. In cell culture the synthetic viruses displayed phenotypes comparable to the natural isolate, and in a mouse model they caused lethal infections that were indistinguishable from infections with a natural strain. Compared to ITA07-RA1 and clinical isolate NL10/152, the synthetic viruses displayed similar sensitivities to several antiviral compounds. 3-deaza-adenosine was identified as a new inhibitor of CHIKV replication. Cyclosporin A had no effect on CHIKV replication, suggesting that cyclophilins -opposite to what was found for other +RNA viruses- do not play an essential role in CHIKV replication. The characterization of the consensus sequence-based synthetic viruses and their comparison to natural isolates demonstrated that CHIKV LS3 and LS3-GFP are suitable and representative tools to study CHIKV-host interactions, screen for antiviral compounds and unravel their mode of action.


Assuntos
Vírus Chikungunya/genética , Vírus Chikungunya/isolamento & purificação , Sequência Consenso , Biologia Sintética , Animais , Anticorpos Monoclonais/imunologia , Antivirais/farmacologia , Linhagem Celular , Vírus Chikungunya/efeitos dos fármacos , Vírus Chikungunya/fisiologia , DNA Complementar/genética , Cinética , Camundongos , RNA de Cadeia Dupla/biossíntese , RNA de Cadeia Dupla/metabolismo , RNA Viral/biossíntese , RNA Viral/metabolismo , Proteínas Virais/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA