Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 298(6): 101956, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35452674

RESUMEN

The signaling pathways and cellular functions regulated by the four Numb-associated kinases are largely unknown. We reported that AAK1 and GAK control intracellular trafficking of RNA viruses and revealed a requirement for BIKE in early and late stages of dengue virus (DENV) infection. However, the downstream targets phosphorylated by BIKE have not yet been identified. Here, to identify BIKE substrates, we conducted a barcode fusion genetics-yeast two-hybrid screen and retrieved publicly available data generated via affinity-purification mass spectrometry. We subsequently validated 19 of 47 putative BIKE interactors using mammalian cell-based protein-protein interaction assays. We found that CLINT1, a cargo-specific adapter implicated in bidirectional Golgi-to-endosome trafficking, emerged as a predominant hit in both screens. Our experiments indicated that BIKE catalyzes phosphorylation of a threonine 294 CLINT1 residue both in vitro and in cell culture. Our findings revealed that CLINT1 phosphorylation mediates its binding to the DENV nonstructural 3 protein and subsequently promotes DENV assembly and egress. Additionally, using live-cell imaging we revealed that CLINT1 cotraffics with DENV particles and is involved in mediating BIKE's role in DENV infection. Finally, our data suggest that additional cellular BIKE interactors implicated in the host immune and stress responses and the ubiquitin proteasome system might also be candidate phosphorylation substrates of BIKE. In conclusion, these findings reveal cellular substrates and pathways regulated by the understudied Numb-associated kinase enzyme BIKE, a mechanism for CLINT1 regulation, and control of DENV infection via BIKE signaling, with potential implications for cell biology, virology, and host-targeted antiviral design.


Asunto(s)
Virus del Dengue , Dengue , Animales , Dengue/metabolismo , Virus del Dengue/metabolismo , Humanos , Fosforilación , Técnicas del Sistema de Dos Híbridos , Replicación Viral
2.
Antiviral Res ; 184: 104966, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33137362

RESUMEN

Global health is threatened by emerging viruses, many of which lack approved therapies and effective vaccines, including dengue, Ebola, and Venezuelan equine encephalitis. We previously reported that AAK1 and GAK, two of the four members of the understudied Numb-associated kinases (NAK) family, control intracellular trafficking of RNA viruses. Nevertheless, the role of BIKE and STK16 in viral infection remained unknown. Here, we reveal a requirement for BIKE, but not STK-16, in dengue virus (DENV) infection. BIKE mediates both early (postinternalization) and late (assembly/egress) stages in the DENV life cycle, and this effect is mediated in part by phosphorylation of a threonine 156 (T156) residue in the µ subunit of the adaptor protein (AP) 2 complex. Pharmacological compounds with potent anti-BIKE activity, including the investigational anticancer drug 5Z-7-oxozeaenol and more selective inhibitors, suppress DENV infection both in vitro and ex vivo. BIKE overexpression reverses the antiviral activity, validating that the mechanism of antiviral action is, at least in part, mediated by BIKE. Lastly, 5Z-7-oxozeaenol exhibits antiviral activity against viruses from three unrelated RNA viral families with a high genetic barrier to resistance. These findings reveal regulation of poorly understood stages of the DENV life cycle via BIKE signaling and establish a proof-of-principle that pharmacological inhibition of BIKE can be potentially used as a broad-spectrum strategy against acute emerging viral infections.


Asunto(s)
Virus del Dengue/fisiología , Dengue/virología , Lactonas/farmacología , Proteínas Serina-Treonina Quinasas/fisiología , Resorcinoles/farmacología , Factores de Transcripción/fisiología , Proteínas Adaptadoras del Transporte Vesicular/antagonistas & inhibidores , Animales , Antivirales/farmacología , Línea Celular , Chlorocebus aethiops , Dengue/tratamiento farmacológico , Virus del Dengue/efectos de los fármacos , Reposicionamiento de Medicamentos , Interacciones Microbiota-Huesped , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , ARN Viral , Proteínas Recombinantes , Transducción de Señal , Células Vero , Internalización del Virus , Replicación Viral
3.
Cell Rep ; 26(7): 1800-1814.e5, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30759391

RESUMEN

The mechanisms that regulate envelopment of HCV and other viruses that bud intracellularly and/or lack late-domain motifs are largely unknown. We reported that K63 polyubiquitination of the HCV nonstructural (NS) 2 protein mediates HRS (ESCRT-0 component) binding and envelopment. Nevertheless, the ubiquitin signaling that governs NS2 ubiquitination remained unknown. Here, we map the NS2 interactome with the ubiquitin proteasome system (UPS) via mammalian cell-based screens. NS2 interacts with E3 ligases, deubiquitinases, and ligase regulators, some of which are candidate proviral or antiviral factors. MARCH8, a RING-finger E3 ligase, catalyzes K63-linked NS2 polyubiquitination in vitro and in HCV-infected cells. MARCH8 is required for infection with HCV, dengue, and Zika viruses and specifically mediates HCV envelopment. Our data reveal regulation of HCV envelopment via ubiquitin signaling and both a viral protein substrate and a ubiquitin K63-linkage of the understudied MARCH8, with potential implications for cell biology, virology, and host-targeted antiviral design.


Asunto(s)
Hepacivirus/metabolismo , Hepatitis C/virología , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Células HEK293 , Hepacivirus/patogenicidad , Hepatitis C/genética , Hepatitis C/metabolismo , Humanos , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
4.
Cell Mol Life Sci ; 75(20): 3693-3714, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30043139

RESUMEN

Viruses are obligate intracellular pathogens that are dependent on cellular machineries for their replication. Recent technological breakthroughs have facilitated reliable identification of host factors required for viral infections and better characterization of the virus-host interplay. While these studies have revealed cellular machineries that are uniquely required by individual viruses, accumulating data also indicate the presence of broadly required mechanisms. Among these overlapping cellular functions are components of intracellular membrane trafficking pathways. Here, we review recent discoveries focused on how viruses exploit intracellular membrane trafficking pathways to promote various stages of their life cycle, with an emphasis on cellular factors that are usurped by a broad range of viruses. We describe broadly required components of the endocytic and secretory pathways, the Endosomal Sorting Complexes Required for Transport pathway, and the autophagy pathway. Identification of such overlapping host functions offers new opportunities to develop broad-spectrum host-targeted antiviral strategies.


Asunto(s)
Membranas Intracelulares/metabolismo , Virus/metabolismo , Autofagia , Clatrina/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Interacciones Huésped-Patógeno , Humanos , Membranas Intracelulares/virología , Lisosomas/metabolismo , Vías Secretoras , Internalización del Virus , Virus/patogenicidad
5.
J Med Chem ; 61(14): 6178-6192, 2018 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-29953812

RESUMEN

There is an urgent need for strategies to combat dengue and other emerging viral infections. We reported that cyclin G-associated kinase (GAK), a cellular regulator of the clathrin-associated host adaptor proteins AP-1 and AP-2, regulates intracellular trafficking of multiple unrelated RNA viruses during early and late stages of the viral lifecycle. We also reported the discovery of potent, selective GAK inhibitors based on an isothiazolo[4,3- b]pyridine scaffold, albeit with moderate antiviral activity. Here, we describe our efforts leading to the discovery of novel isothiazolo[4,3- b]pyridines that maintain high GAK affinity and selectivity. These compounds demonstrate improved in vitro activity against dengue virus, including in human primary dendritic cells, and efficacy against the unrelated Ebola and chikungunya viruses. Moreover, inhibition of GAK activity was validated as an important mechanism of antiviral action of these compounds. These findings demonstrate the potential utility of a GAK-targeted broad-spectrum approach for combating currently untreatable emerging viral infections.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridinas/química , Piridinas/farmacología , Tiazoles/química , Antivirales/química , Antivirales/farmacología , Línea Celular , Humanos , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
6.
Antiviral Res ; 155: 67-75, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29753658

RESUMEN

There is an urgent need for strategies to combat dengue virus (DENV) infection; a major global threat. We reported that the cellular kinases AAK1 and GAK regulate intracellular trafficking of multiple viruses and that sunitinib and erlotinib, approved anticancer drugs with potent activity against these kinases, protect DENV-infected mice from mortality. Nevertheless, further characterization of the therapeutic potential and underlying mechanism of this approach is required prior to clinical evaluation. Here, we demonstrate that sunitinib/erlotinib combination achieves sustained suppression of systemic infection at approved dose in DENV-infected IFN-α/ß and IFN-γ receptor-deficient mice. Nevertheless, treatment with these blood-brain barrier impermeable drugs delays, yet does not prevent, late-onset paralysis; a common manifestation in this immunodeficient mouse model but not in humans. Sunitinib and erlotinib treatment also demonstrates efficacy in human primary monocyte-derived dendritic cells. Additionally, DENV infection induces expression of AAK1 transcripts, but not GAK, via single-cell transcriptomics, and these kinases are important molecular targets underlying the anti-DENV effect of sunitinib and erlotinib. Lastly, sunitinib/erlotinib combination alters inflammatory cytokine responses in DENV-infected mice. These findings support feasibility of repurposing sunitinib/erlotinib combination as a host-targeted antiviral approach and contribute to understanding its mechanism of antiviral action.


Asunto(s)
Antivirales/uso terapéutico , Virus del Dengue/efectos de los fármacos , Dengue/tratamiento farmacológico , Reposicionamiento de Medicamentos , Clorhidrato de Erlotinib/uso terapéutico , Sunitinib/uso terapéutico , Animales , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/virología , Virus del Dengue/fisiología , Estudios de Factibilidad , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Proteínas Serina-Treonina Quinasas/genética , Análisis de la Célula Individual , Replicación Viral/efectos de los fármacos
7.
mBio ; 9(2)2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29535204

RESUMEN

Hepatitis C virus (HCV) spreads via secreted cell-free particles or direct cell-to-cell transmission. Yet, virus-host determinants governing differential intracellular trafficking of cell-free- and cell-to-cell-transmitted virus remain unknown. The host adaptor proteins (APs) AP-1A, AP-1B, and AP-4 traffic in post-Golgi compartments, and the latter two are implicated in basolateral sorting. We reported that AP-1A mediates HCV trafficking during release, whereas the endocytic adaptor AP-2 mediates entry and assembly. We demonstrated that the host kinases AAK1 and GAK regulate HCV infection by controlling these clathrin-associated APs. Here, we sought to define the roles of AP-4, a clathrin-independent adaptor; AP-1A; and AP-1B in HCV infection. We screened for interactions between HCV proteins and the µ subunits of AP-1A, AP-1B, and AP-4 by mammalian cell-based protein fragment complementation assays. The nonstructural 2 (NS2) protein emerged as an interactor of these adaptors in this screening and by coimmunoprecipitations in HCV-infected cells. Two previously unrecognized dileucine-based motifs in the NS2 C terminus mediated AP binding and HCV release. Infectivity and coculture assays demonstrated that while all three adaptors mediate HCV release and cell-free spread, AP-1B and AP-4, but not AP-1A, mediate cell-to-cell spread. Live-cell imaging revealed HCV cotrafficking with AP-1A, AP-1B, and AP-4 and that AP-4 mediates HCV trafficking in a post-Golgi compartment. Lastly, HCV cell-to-cell spread was regulated by AAK1 and GAK and thus susceptible to treatment with AAK1 and GAK inhibitors. These data provide a mechanistic understanding of HCV trafficking in distinct release pathways and reveal a requirement for APs in cell-to-cell viral spread.IMPORTANCE HCV spreads via cell-free infection or cell-to-cell contact that shields it from antibody neutralization, thereby facilitating viral persistence. Yet, factors governing this differential sorting remain unknown. By integrating proteomic, RNA interference, genetic, live-cell imaging, and pharmacological approaches, we uncover differential coopting of host adaptor proteins (APs) to mediate HCV traffic at distinct late steps of the viral life cycle. We reported that AP-1A and AP-2 mediate HCV trafficking during release and assembly, respectively. Here, we demonstrate that dileucine motifs in the NS2 protein mediate AP-1A, AP-1B, and AP-4 binding and cell-free virus release. Moreover, we reveal that AP-4, an adaptor not previously implicated in viral infections, mediates cell-to-cell spread and HCV trafficking. Lastly, we demonstrate cell-to-cell spread regulation by AAK1 and GAK, host kinases controlling APs, and susceptibility to their inhibitors. This study provides mechanistic insights into virus-host determinants that facilitate HCV trafficking, with potential implications for pathogenesis and antiviral agent design.


Asunto(s)
Complejo 1 de Proteína Adaptadora/metabolismo , Complejo 4 de Proteína Adaptadora/metabolismo , Hepacivirus/fisiología , Interacciones Huésped-Patógeno , Proteínas no Estructurales Virales/metabolismo , Liberación del Virus , Línea Celular , Humanos , Inmunoprecipitación , Unión Proteica , Mapeo de Interacción de Proteínas
9.
ACS Infect Dis ; 4(2): 88-92, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29298032

RESUMEN

There is a large, global unmet need for the development of countermeasures to combat intracellular pathogens. The development of novel antimicrobials is expensive and slow and typically focuses on selective inhibition of proteins encoded by a single pathogen, thereby providing a narrow spectrum of coverage. The repurposing of approved drugs targeting host functions required for microbial infections represents a promising alternative. This review summarizes progress and challenges in the repurposing of approved drugs as host-targeted broad-spectrum agents for the treatment of intracellular pathogens. These strategies include targeting both cellular factors required for infection by various viruses, intracellular bacteria, and/or protozoa as well as factors that modulate the host immune response to these microbial infections. The repurposed approach offers complementary means to develop therapeutics against existing and emerging intracellular microbial threats.


Asunto(s)
Antiinfecciosos/farmacología , Reposicionamiento de Medicamentos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/microbiología , Animales , Antineoplásicos/farmacología , Humanos , Espacio Intracelular/virología , Terapia Molecular Dirigida
10.
DNA Cell Biol ; 37(2): 63-69, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29148875

RESUMEN

The high cost of drug development and the narrow spectrum of coverage typically provided by direct-acting antivirals limit the scalability of this antiviral approach. This review summarizes progress and challenges in the repurposing of approved kinase inhibitors as host-targeted broad-spectrum antiviral therapies.


Asunto(s)
Antivirales/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antivirales/uso terapéutico , Reposicionamiento de Medicamentos , Humanos , Sistema de Señalización de MAP Quinasas , Inhibidores de Proteínas Quinasas/uso terapéutico , Virosis/tratamiento farmacológico , Virosis/enzimología
11.
mBio ; 7(6)2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27803188

RESUMEN

Enveloped viruses commonly utilize late-domain motifs, sometimes cooperatively with ubiquitin, to hijack the endosomal sorting complex required for transport (ESCRT) machinery for budding at the plasma membrane. However, the mechanisms underlying budding of viruses lacking defined late-domain motifs and budding into intracellular compartments are poorly characterized. Here, we map a network of hepatitis C virus (HCV) protein interactions with the ESCRT machinery using a mammalian-cell-based protein interaction screen and reveal nine novel interactions. We identify HRS (hepatocyte growth factor-regulated tyrosine kinase substrate), an ESCRT-0 complex component, as an important entry point for HCV into the ESCRT pathway and validate its interactions with the HCV nonstructural (NS) proteins NS2 and NS5A in HCV-infected cells. Infectivity assays indicate that HRS is an important factor for efficient HCV assembly. Specifically, by integrating capsid oligomerization assays, biophysical analysis of intracellular viral particles by continuous gradient centrifugations, proteolytic digestion protection, and RNase digestion protection assays, we show that HCV co-opts HRS to mediate a late assembly step, namely, envelopment. In the absence of defined late-domain motifs, K63-linked polyubiquitinated lysine residues in the HCV NS2 protein bind the HRS ubiquitin-interacting motif to facilitate assembly. Finally, ESCRT-III and VPS/VTA1 components are also recruited by HCV proteins to mediate assembly. These data uncover involvement of ESCRT proteins in intracellular budding of a virus lacking defined late-domain motifs and a novel mechanism by which HCV gains entry into the ESCRT network, with potential implications for other viruses. IMPORTANCE: Viruses commonly bud at the plasma membrane by recruiting the host ESCRT machinery via conserved motifs termed late domains. The mechanism by which some viruses, such as HCV, bud intracellularly is, however, poorly characterized. Moreover, whether envelopment of HCV and other viruses lacking defined late domains is ESCRT mediated and, if so, what the entry points into the ESCRT pathway are remain unknown. Here, we report the interaction network of HCV with the ESCRT machinery and a critical role for HRS, an ESCRT-0 complex component, in HCV envelopment. Viral protein ubiquitination was discovered to be a signal for HRS binding and HCV assembly, thereby functionally compensating for the absence of late domains. These findings characterize how a virus lacking defined late domains co-opts ESCRT to bud intracellularly. Since the ESCRT machinery is essential for the life cycle of multiple viruses, better understanding of this virus-host interplay may yield targets for broad-spectrum antiviral therapies.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Hepacivirus/fisiología , Interacciones Huésped-Patógeno , Proteínas no Estructurales Virales/metabolismo , Ensamble de Virus , Liberación del Virus , Mapeo de Interacción de Proteínas , Ubiquitinación
12.
J Mol Biol ; 427(1): 205-20, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24979679

RESUMEN

Proton-pumping rhodopsins (PPRs) are photoactive retinal-binding proteins that transport ions across biological membranes in response to light. These proteins are interesting for light-harvesting applications in bioenergy production, in optogenetics applications in neuroscience, and as fluorescent sensors of membrane potential. Little is known, however, about how the protein sequence determines the considerable variation in spectral properties of PPRs from different biological niches or how to engineer these properties in a given PPR. Here we report a comprehensive study of amino acid substitutions in the retinal-binding pocket of Gloeobacter violaceus rhodopsin (GR) that tune its spectral properties. Directed evolution generated 70 GR variants with absorption maxima shifted by up to ±80nm, extending the protein's light absorption significantly beyond the range of known natural PPRs. While proton-pumping activity was disrupted in many of the spectrally shifted variants, we identified single tuning mutations that incurred blue and red shifts of 42nm and 22nm, respectively, that did not disrupt proton pumping. Blue-shifting mutations were distributed evenly along the retinal molecule while red-shifting mutations were clustered near the residue K257, which forms a covalent bond with retinal through a Schiff base linkage. Thirty eight of the identified tuning mutations are not found in known microbial rhodopsins. We discovered a subset of red-shifted GRs that exhibit high levels of fluorescence relative to the WT (wild-type) protein.


Asunto(s)
Cianobacterias/metabolismo , Evolución Molecular Dirigida , Bombas de Protones/metabolismo , Rodopsinas Microbianas/química , Rodopsinas Microbianas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Cianobacterias/genética , Cianobacterias/crecimiento & desarrollo , Fluorescencia , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Rodopsinas Microbianas/genética , Homología de Secuencia de Aminoácido
13.
ACS Chem Biol ; 6(5): 502-10, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21294551

RESUMEN

Although protein kinases have recently emerged as important drug targets, the anti-infective potential of protein kinase inhibitors has not been developed extensively. We identified the mammalian PDK1 inhibitor KP-372-1 as a potent antifungal molecule with activity against yeast and fungal biofilms using a screening strategy for protein kinase inhibitors that block the cell wall stress response in yeast. Genetic and biochemical studies indicate that KP-372-1 inhibits fungal PDK1 orthologs (Pkh kinases) as part of its mode of action and support a role for Pkh kinases in eisosome assembly. Two other structurally distinct molecules that inhibit PDK1, OSU-03012 and UCN-01, also have antifungal activity. Taken together, these data indicate that fungal PDK1 orthologs are promising targets for new antifungal drug development.


Asunto(s)
Antifúngicos/aislamiento & purificación , Inhibidores de Proteínas Quinasas/aislamiento & purificación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Antifúngicos/farmacología , Candida albicans/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Pirazoles/aislamiento & purificación , Pirazoles/farmacología , Saccharomyces cerevisiae/efectos de los fármacos , Estaurosporina/análogos & derivados , Estaurosporina/aislamiento & purificación , Estaurosporina/farmacología , Sulfonamidas/aislamiento & purificación , Sulfonamidas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA