Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Nat Commun ; 15(1): 4153, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755212

RESUMEN

Viral myocarditis, an inflammatory disease of the heart, causes significant morbidity and mortality. Type I interferon (IFN)-mediated antiviral responses protect against myocarditis, but the mechanisms are poorly understood. We previously identified A Disintegrin And Metalloproteinase domain 9 (ADAM9) as an important factor in viral pathogenesis. ADAM9 is implicated in a range of human diseases, including inflammatory diseases; however, its role in viral infection is unknown. Here, we demonstrate that mice lacking ADAM9 are more susceptible to encephalomyocarditis virus (EMCV)-induced death and fail to mount a characteristic type I IFN response. This defect in type I IFN induction is specific to positive-sense, single-stranded RNA (+ ssRNA) viruses and involves melanoma differentiation-associated protein 5 (MDA5)-a key receptor for +ssRNA viruses. Mechanistically, ADAM9 binds to MDA5 and promotes its oligomerization and thereby downstream mitochondrial antiviral-signaling protein (MAVS) activation in response to EMCV RNA stimulation. Our findings identify a role for ADAM9 in the innate antiviral response, specifically MDA5-mediated IFN production, which protects against virus-induced cardiac damage, and provide a potential therapeutic target for treatment of viral myocarditis.


Asunto(s)
Proteínas ADAM , Infecciones por Cardiovirus , Virus de la Encefalomiocarditis , Inmunidad Innata , Interferón Tipo I , Helicasa Inducida por Interferón IFIH1 , Proteínas de la Membrana , Ratones Noqueados , Miocarditis , Animales , Virus de la Encefalomiocarditis/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/inmunología , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/virología , Proteínas ADAM/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/inmunología , Ratones , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Miocarditis/inmunología , Miocarditis/virología , Humanos , Ratones Endogámicos C57BL , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Transducción de Señal/inmunología , Masculino , Células HEK293
2.
Methods Mol Biol ; 2142: 215-234, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32367370

RESUMEN

The discovery and implementation of CRISPR/Cas9 tools in pooled genetic screens have allowed for the rapid, high-fidelity identification of host-virus interactions. However, pooled CRISPR screening has significant limitations in its ability both to perform cell biology and plate reader-based screens and to find alleles that result in intermediate-strength phenotypes. Here we introduce an arrayed CRISPR screening method, FACS-IT, which allows researchers to use high content imaging analysis, plate reader assays, cell supernatant characterization, and percent infectivity to characterize CRISPR-mediated gene disruptions causing both moderate and extreme phenotypic changes. By using flow sorting capabilities and CRISPR libraries that are widely available, FACS-IT overcomes both the significant limitation of pooled screening approaches and the prohibitive costs of large-scale arrayed CRISPR reagents. In doing so, FACS-IT will enable researchers to creatively use CRISPR screening to obtain a deeper understanding of biology across a wide range of fields and applications.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Pruebas Genéticas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Infección por el Virus Zika/diagnóstico , Virus Zika/genética , Virus Zika/patogenicidad , Células A549 , Proteína 9 Asociada a CRISPR/metabolismo , Técnicas de Cultivo de Célula/métodos , Citometría de Flujo , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Fenotipo , ARN Guía de Kinetoplastida/genética , Carga Viral/métodos , Factores de Virulencia/genética , Factores de Virulencia/aislamiento & purificación , Cultivo de Virus/métodos , Virus Zika/fisiología , Infección por el Virus Zika/genética , Infección por el Virus Zika/patología
3.
J Immunol ; 203(7): 1999-2010, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31471524

RESUMEN

To arise and progress, cancers need to evade immune elimination. Consequently, progressing tumors are often MHC class I (MHC-I) low and express immune inhibitory molecules, such as PD-L1, which allows them to avoid the main antitumor host defense, CD8+ T cells. The molecular mechanisms that led to these alterations were incompletely understood. In this study, we identify loss of the transcription factor IRF2 as a frequent underlying mechanism that leads to a tumor immune evasion phenotype in both humans and mice. We identified IRF2 in a CRISPR-based forward genetic screen for genes that controlled MHC-I Ag presentation in HeLa cells. We then found that many primary human cancers, including lung, colon, breast, prostate, and others, frequently downregulated IRF2. Although IRF2 is generally known as a transcriptional repressor, we found that it was a transcriptional activator of many key components of the MHC-I pathway, including immunoproteasomes, TAP, and ERAP1, whose transcriptional control was previously poorly understood. Upon loss of IRF2, cytosol-to-endoplasmic reticulum peptide transport and N-terminal peptide trimming become rate limiting for Ag presentation. In addition, we found that IRF2 is a repressor of PD-L1. Thus, by downregulating a single nonessential gene, tumors become harder to see (reduced Ag presentation), more inhibitory (increased checkpoint inhibitor), and less susceptible to being killed by CD8+ T cells. Importantly, we found that the loss of Ag presentation caused by IRF2 downregulation could be reversed by IFN-stimulated induction of the transcription factor IRF1. The implication of these findings for tumor progression and immunotherapy are discussed.


Asunto(s)
Presentación de Antígeno , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Factor 2 Regulador del Interferón/deficiencia , Proteínas de Neoplasias/inmunología , Neoplasias , Escape del Tumor , Antígeno B7-H1/genética , Linfocitos T CD8-positivos/patología , Regulación hacia Abajo/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Células HEK293 , Células HeLa , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Factor 2 Regulador del Interferón/inmunología , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología
4.
Nat Med ; 25(9): 1377-1384, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31501601

RESUMEN

People living with HIV (PLWH) have expressed concern about the life-long burden and stigma associated with taking pills daily and can experience medication fatigue that might lead to suboptimal treatment adherence and the emergence of drug-resistant viral variants, thereby limiting future treatment options1-3. As such, there is strong interest in long-acting antiretroviral (ARV) agents that can be administered less frequently4. Herein, we report GS-CA1, a new archetypal small-molecule HIV capsid inhibitor with exceptional potency against HIV-2 and all major HIV-1 types, including viral variants resistant to the ARVs currently in clinical use. Mechanism-of-action studies indicate that GS-CA1 binds directly to the HIV-1 capsid and interferes with capsid-mediated nuclear import of viral DNA, HIV particle production and ordered capsid assembly. GS-CA1 selects in vitro for unfit GS-CA1-resistant capsid variants that remain fully susceptible to other classes of ARVs. Its high metabolic stability and low solubility enabled sustained drug release in mice following a single subcutaneous dosing. GS-CA1 showed high antiviral efficacy as a long-acting injectable monotherapy in a humanized mouse model of HIV-1 infection, outperforming long-acting rilpivirine. Collectively, these results demonstrate the potential of ultrapotent capsid inhibitors as new long-acting agents for the treatment of HIV-1 infection.


Asunto(s)
Fármacos Anti-VIH/farmacología , Proteínas de la Cápside/antagonistas & inhibidores , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Indazoles/farmacología , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Fármacos Anti-VIH/uso terapéutico , Cápside/efectos de los fármacos , Cápside/metabolismo , Proteínas de la Cápside/genética , ADN Viral/efectos de los fármacos , Preparaciones de Acción Retardada , Farmacorresistencia Viral/efectos de los fármacos , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/genética , VIH-1/patogenicidad , VIH-2/efectos de los fármacos , VIH-2/patogenicidad , Humanos , Indazoles/uso terapéutico , Cumplimiento de la Medicación , Ratones , Piridinas/uso terapéutico
5.
J Life Sci (Westlake Village) ; 1(1): 39-45, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31448372

RESUMEN

HIV-1 integration favors active chromatin, which is primarily mediated through interactions between the viral capsid and integrase proteins with host factors cleavage and polyadenylation specificity factor 6 (CPSF6) and lens epithelium-derived growth factor/p75, respectively. Previously published image-based studies had suggested that HIV-1 prefers to integrate into chromatin that associates spatially with the nuclear periphery. Here, we re-evaluated previously reported HIV-1 nuclear distance measures across studies and show that HIV-1 prefers peri-nuclear and mid-nuclear zones similarly, with a common preference between studies mapping to the boundary between these two radial areas. We also discuss emerging roles for the capsid-CPSF6 interaction in facilitating HIV-1 pre-integration complex nuclear import and subsequent intranuclear trafficking to preferred sites of viral DNA integration.

6.
Proc Natl Acad Sci U S A ; 116(14): 7043-7052, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30894498

RESUMEN

A human cytomegalovirus (HCMV) pentameric glycoprotein complex (PC), gH-gL-UL128-UL130-UL131A, is necessary for viral infection of clinically relevant cell types, including epithelial cells, which are important for interhost transmission and disease. We performed genome-wide CRISPR/Cas9 screens of different cell types in parallel to identify host genes specifically required for HCMV infection of epithelial cells. This effort identified a multipass membrane protein, OR14I1, as a receptor for HCMV infection. This olfactory receptor family member is required for HCMV attachment, entry, and infection of epithelial cells and is dependent on the presence of viral PC. OR14I1 is required for AKT activation and mediates endocytosis entry of HCMV. We further found that HCMV infection of epithelial cells is blocked by a synthetic OR14I1 peptide and inhibitors of adenylate cyclase and protein kinase A (PKA) signaling. Identification of OR14I1 as a PC-dependent HCMV host receptor associated with epithelial tropism and the role of the adenylate cyclase/PKA/AKT-mediated signaling pathway in HCMV infection reveal previously unappreciated targets for the development of vaccines and antiviral therapies.


Asunto(s)
Citomegalovirus/fisiología , Células Epiteliales/metabolismo , Complejos Multiproteicos/metabolismo , Transducción de Señal , Proteínas Virales/metabolismo , Tropismo Viral/fisiología , Células A549 , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Células HEK293 , Células HeLa , Humanos , Complejos Multiproteicos/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Virales/genética
7.
mBio ; 10(1)2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30723129

RESUMEN

Encephalomyocarditis virus (EMCV) is a picornavirus that produces lytic infections in murine and human cells. Employing a genome-wide CRISPR-Cas9 knockout screen to find host factors required for EMCV infection, we identified a role for ADAM9 in EMCV infection. CRISPR-mediated deletion of ADAM9 in multiple human cell lines rendered the cells highly resistant to EMCV infection and cell death. Primary fibroblasts from ADAM9 KO mice were also strongly resistant to EMCV infection and cell death. In contrast, ADAM9 KO and WT cells were equally susceptible to infection with other viruses, including the picornavirus Coxsackie virus B. ADAM9 KO cells failed to produce viral progeny when incubated with EMCV. However, bypassing EMCV entry into cells through delivery of viral RNA directly to the cytosol yielded infectious EMCV virions from ADAM9 KO cells, suggesting that ADAM9 is not required for EMCV replication post-entry. These findings establish that ADAM9 is required for the early stage of EMCV infection, likely for virus entry or viral genome delivery to the cytosol.IMPORTANCE Viral myocarditis is a leading cause of death in the United States, contributing to numerous unexplained deaths in people ≤35 years old. Enteroviruses contribute to many cases of human myocarditis. Encephalomyocarditis virus (EMCV) infection causes viral myocarditis in rodent models, but its receptor requirements have not been fully identified. CRISPR-Cas9 screens can identify host dependency factors essential for EMCV infection and enhance our understanding of key events that follow viral infection, potentially leading to new strategies for preventing viral myocarditis. Using a CRISPR-Cas9 screen, we identified adisintegrin and metalloproteinase 9 domain (ADAM9) as a major factor required for the early stages of EMCV infection in both human and murine infection.


Asunto(s)
Proteínas ADAM/metabolismo , Infecciones por Cardiovirus/genética , Resistencia a la Enfermedad , Virus de la Encefalomiocarditis/crecimiento & desarrollo , Proteínas de la Membrana/metabolismo , Animales , Línea Celular , Técnicas de Inactivación de Genes , Pruebas Genéticas , Humanos , Ratones , Ratones Noqueados , Modelos Biológicos
8.
iScience ; 12: 270-279, 2019 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-30716700

RESUMEN

Congenital microcephaly occurs in utero during Zika virus (ZIKV) infection. The single-gene disorder, Majewski osteodysplastic primordial dwarfism type II (MOPDII), also leads to microcephaly and is concomitant with a decrease in the centrosomal protein, pericentrin (PCNT). This protein is a known contributor of mitotic spindle misorientation and ultimately, microcephaly. Similar to MOPDII, either viral infection or interferon (IFN)-α exposure reduced PCNT levels at the mitotic spindle poles. We unexpectedly found that infection of cells with any one of a diverse set of viruses, such as ZIKV, dengue virus, cytomegalovirus, influenza A virus, or hepatitis B virus, or treatment of cells with the anti-viral cytokine, IFN-α, produced mitotic spindle misorientation. These findings demonstrate a related mechanism for the development of microcephaly in viral infection, the host's antiviral IFN response, and primordial dwarfism.

9.
PLoS Pathog ; 15(1): e1007532, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30640957

RESUMEN

Late endosome-resident interferon-induced transmembrane protein 3 (IFITM3) inhibits fusion of diverse viruses, including Influenza A virus (IAV), by a poorly understood mechanism. Despite the broad antiviral activity of IFITM3, viruses like Lassa virus (LASV), are fully resistant to its inhibitory effects. It is currently unclear whether resistance arises from a highly efficient fusion machinery that is capable of overcoming IFITM3 restriction or the ability to enter from cellular sites devoid of this factor. Here, we constructed and validated a functional IFITM3 tagged with EGFP or other fluorescent proteins. This breakthrough allowed live cell imaging of virus co-trafficking and fusion with endosomal compartments in cells expressing fluorescent IFITM3. Three-color single virus and endosome tracking revealed that sensitive (IAV), but not resistant (LASV), viruses become trapped within IFITM3-positive endosomes where they underwent hemifusion but failed to release their content into the cytoplasm. IAV fusion with IFITM3-containing compartments could be rescued by amphotericin B treatment, which has been previously shown to antagonize the antiviral activity of this protein. By comparison, virtually all LASV particles trafficked and fused with endosomes lacking detectable levels of fluorescent IFITM3, implying that this virus escapes restriction by utilizing endocytic pathways that are distinct from the IAV entry pathways. The importance of virus uptake and transport pathways is further reinforced by the observation that LASV glycoprotein-mediated cell-cell fusion is inhibited by IFITM3 and other members of the IFITM family expressed in target cells. Together, our results strongly support a model according to which IFITM3 accumulation at the sites of virus fusion is a prerequisite for its antiviral activity and that this protein traps viral fusion at a hemifusion stage by preventing the formation of fusion pores. We conclude that the ability to utilize alternative endocytic pathways for entry confers IFITM3-resistance to otherwise sensitive viruses.


Asunto(s)
Endosomas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/fisiología , Células A549/metabolismo , Animales , Antivirales/metabolismo , Células COS/metabolismo , Chlorocebus aethiops , Endosomas/virología , Células HEK293/metabolismo , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/patogenicidad , Interferones/metabolismo , Virus Lassa/patogenicidad , Imagen Óptica/métodos , Transporte de Proteínas , Internalización del Virus
10.
Cell Host Microbe ; 24(3): 392-404.e8, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30173955

RESUMEN

HIV-1 integration into the host genome favors actively transcribed genes. Prior work indicated that the nuclear periphery provides the architectural basis for integration site selection, with viral capsid-binding host cofactor CPSF6 and viral integrase-binding cofactor LEDGF/p75 contributing to selection of individual sites. Here, by investigating the early phase of infection, we determine that HIV-1 traffics throughout the nucleus for integration. CPSF6-capsid interactions allow the virus to bypass peripheral heterochromatin and penetrate the nuclear structure for integration. Loss of interaction with CPSF6 dramatically alters virus localization toward the nuclear periphery and integration into transcriptionally repressed lamina-associated heterochromatin, while loss of LEDGF/p75 does not significantly affect intranuclear HIV-1 localization. Thus, CPSF6 serves as a master regulator of HIV-1 intranuclear localization by trafficking viral preintegration complexes away from heterochromatin at the periphery toward gene-dense chromosomal regions within the nuclear interior.


Asunto(s)
Cápside/metabolismo , Núcleo Celular/virología , ADN Viral/genética , Infecciones por VIH/metabolismo , VIH-1/fisiología , Integración Viral , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , ADN Viral/metabolismo , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/genética , Interacciones Huésped-Patógeno , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Unión Proteica , Replicación Viral , Factores de Escisión y Poliadenilación de ARNm/genética
11.
PLoS Comput Biol ; 14(4): e1006093, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29677181

RESUMEN

Mounting evidence suggests that glycans, rather than merely serving as a "shield", contribute critically to antigenicity of the HIV envelope (Env) glycoprotein, representing critical antigenic determinants for many broadly neutralizing antibodies (bNAbs). While many studies have focused on defining the role of individual glycans or groups of proximal glycans in bNAb binding, little is known about the effects of changes in the overall glycan landscape in modulating antibody access and Env antigenicity. Here we developed a systems glycobiology approach to reverse engineer the complexity of HIV glycan heterogeneity to guide antigenicity-based de novo glycoprotein design. bNAb binding was assessed against a panel of 94 recombinant gp120 monomers exhibiting defined glycan site occupancies. Using a Bayesian machine learning algorithm, bNAb-specific glycan footprints were identified and used to design antigens that selectively alter bNAb antigenicity as a proof-of concept. Our approach provides a new design strategy to predictively modulate antigenicity via the alteration of glycan topography, thereby focusing the humoral immune response on sites of viral vulnerability for HIV.


Asunto(s)
Antígenos VIH/química , Antígenos VIH/inmunología , Proteína gp120 de Envoltorio del VIH/química , Proteína gp120 de Envoltorio del VIH/inmunología , Polisacáridos/química , Polisacáridos/inmunología , Algoritmos , Secuencia de Aminoácidos , Anticuerpos Neutralizantes , Teorema de Bayes , Sitios de Unión , Biología Computacional , Epítopos/química , Epítopos/genética , Glicosilación , VIH/química , VIH/inmunología , Anticuerpos Anti-VIH , Antígenos VIH/genética , Proteína gp120 de Envoltorio del VIH/genética , Humanos , Aprendizaje Automático , Modelos Moleculares , Ingeniería de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Biología de Sistemas
12.
Curr Opin Virol ; 29: 87-100, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29684735

RESUMEN

Viruses impose an immense burden on human health. With the goal of treating and preventing viral infections, researchers have carried out genetic screens to improve our understanding of viral dependencies and identify potential anti-viral strategies. The emergence of CRISPR genetic screening tools has facilitated this effort by enabling host-virus screens to be undertaken in a more versatile and fidelitous manner than previously possible. Here we review the growing number of CRISPR screens which continue to increase our understanding of host-virus interactions.


Asunto(s)
Sistemas CRISPR-Cas , Pruebas Genéticas , Interacciones Huésped-Patógeno , Virosis/virología , Virus/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Flavivirus/genética , Flavivirus/metabolismo , Humanos , Receptores Virales/metabolismo , Flujo de Trabajo
13.
Nature ; 538(7625): 350-355, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27680706

RESUMEN

Clostridium difficile toxin B (TcdB) is a critical virulence factor that causes diseases associated with C. difficile infection. Here we carried out CRISPR-Cas9-mediated genome-wide screens and identified the members of the Wnt receptor frizzled family (FZDs) as TcdB receptors. TcdB binds to the conserved Wnt-binding site known as the cysteine-rich domain (CRD), with the highest affinity towards FZD1, 2 and 7. TcdB competes with Wnt for binding to FZDs, and its binding blocks Wnt signalling. FZD1/2/7 triple-knockout cells are highly resistant to TcdB, and recombinant FZD2-CRD prevented TcdB binding to the colonic epithelium. Colonic organoids cultured from FZD7-knockout mice, combined with knockdown of FZD1 and 2, showed increased resistance to TcdB. The colonic epithelium in FZD7-knockout mice was less susceptible to TcdB-induced tissue damage in vivo. These findings establish FZDs as physiologically relevant receptors for TcdB in the colonic epithelium.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Colon/metabolismo , Epitelio/metabolismo , Receptores Frizzled/metabolismo , Animales , Antígenos/metabolismo , Proteínas Bacterianas/química , Toxinas Bacterianas/química , Sitios de Unión , Células CHO , Sistemas CRISPR-Cas , Línea Celular , Clostridioides difficile/patogenicidad , Cricetulus , Femenino , Receptores Frizzled/química , Receptores Frizzled/deficiencia , Receptores Frizzled/genética , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Ratones Noqueados , Oligopéptidos/química , Oligopéptidos/metabolismo , Organoides/metabolismo , Dominios Proteicos , Proteoglicanos/metabolismo , Factores de Virulencia/metabolismo , Proteínas Wnt/metabolismo
14.
Cell Rep ; 15(11): 2323-30, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27268505

RESUMEN

Zika virus has emerged as a severe health threat with a rapidly expanding range. The IFITM family of restriction factors inhibits the replication of a broad range of viruses, including the closely related flaviruses West Nile virus and dengue virus. Here, we show that IFITM1 and IFITM3 inhibit Zika virus infection early in the viral life cycle. Moreover, IFITM3 can prevent Zika-virus-induced cell death. These results suggest that strategies to boost the actions and/or levels of the IFITMs might be useful for inhibiting a broad range of emerging viruses.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Replicación Viral/fisiología , Virus Zika/fisiología , Células A549 , Animales , Efecto Citopatogénico Viral , Eliminación de Gen , Sitios Genéticos , Células HeLa , Humanos , Ratones , Transporte de Proteínas , Infección por el Virus Zika
15.
Cell Rep ; 16(1): 232-246, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27342126

RESUMEN

The flaviviruses dengue virus (DENV) and Zika virus (ZIKV) are severe health threats with rapidly expanding ranges. To identify the host cell dependencies of DENV and ZIKV, we completed orthologous functional genomic screens using RNAi and CRISPR/Cas9 approaches. The screens recovered the ZIKV entry factor AXL as well as multiple host factors involved in endocytosis (RAB5C and RABGEF), heparin sulfation (NDST1 and EXT1), and transmembrane protein processing and maturation, including the endoplasmic reticulum membrane complex (EMC). We find that both flaviviruses require the EMC for their early stages of infection. Together, these studies generate a high-confidence, systems-wide view of human-flavivirus interactions and provide insights into the role of the EMC in flavivirus replication.


Asunto(s)
Virus del Dengue/genética , Genómica/métodos , Virus Zika/genética , Sistemas CRISPR-Cas , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Pruebas Genéticas , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Membranas Intracelulares/metabolismo , Unión Proteica , Mapas de Interacción de Proteínas , Interferencia de ARN , Replicación Viral
16.
Adv Virus Res ; 94: 1-51, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26997589

RESUMEN

Over the last several years a wealth of transformative human-virus interaction discoveries have been produced using loss-of-function functional genomics. These insights have greatly expanded our understanding of how human pathogenic viruses exploit our cells to replicate. Two technologies have been at the forefront of this genetic revolution, RNA interference (RNAi) and random retroviral insertional mutagenesis using haploid cell lines (haploid cell screening), with the former technology largely predominating. Now the cutting edge gene editing of the CRISPR/Cas9 system has also been harnessed for large-scale functional genomics and is poised to possibly displace these earlier methods. Here we compare and contrast these three screening approaches for elucidating host-virus interactions, outline their key strengths and weaknesses including a comparison of an arrayed multiple orthologous RNAi reagent screen to a pooled CRISPR/Cas9 human rhinovirus 14-human cell interaction screen, and recount some notable insights made possible by each. We conclude with a brief perspective on what might lie ahead for the fast evolving field of human-virus functional genomics.


Asunto(s)
Sistemas CRISPR-Cas , Genómica/métodos , Haploidia , Interacciones Huésped-Patógeno/genética , Interferencia de ARN , Virus/patogenicidad , Proteínas Bacterianas , Proteína 9 Asociada a CRISPR , Endonucleasas , Técnicas de Inactivación de Genes , Pruebas Genéticas/métodos , Humanos/virología , Mutagénesis Insercional , ARN Interferente Pequeño/genética
17.
Cell Rep ; 13(8): 1717-31, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26586435

RESUMEN

Direct visualization of HIV-1 replication would improve our understanding of the viral life cycle. We adapted established technology and reagents to develop an imaging approach, ViewHIV, which allows evaluation of early HIV-1 replication intermediates, from reverse transcription to integration. These methods permit the simultaneous evaluation of both the capsid protein (CA) and viral DNA genome (vDNA) components of HIV-1 in both the cytosol and nuclei of single cells. ViewHIV is relatively rapid, uses readily available reagents in combination with standard confocal microscopy, and can be done with virtually any HIV-1 strain and permissive cell lines or primary cells. Using ViewHIV, we find that CA enters the nucleus and associates with vDNA in both transformed and primary cells. We also find that CA's interaction with the host polyadenylation factor, CPSF6, enhances nuclear entry and potentiates HIV-1's depth of nuclear invasion, potentially aiding the virus's integration into gene-dense regions.


Asunto(s)
Proteínas de la Cápside/metabolismo , Cápside/metabolismo , VIH-1/genética , Integración Viral/genética , Replicación Viral/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citosol/metabolismo , Replicación del ADN/genética , Genoma Viral/genética , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Células HeLa , Humanos , Transcripción Reversa/genética , Factores de Escisión y Poliadenilación de ARNm/metabolismo
18.
Cell Rep ; 12(5): 850-63, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26212330

RESUMEN

Human rhinovirus (HRV) causes upper respiratory infections and asthma exacerbations. We screened multiple orthologous RNAi reagents and identified host proteins that modulate HRV replication. Here, we show that RNASEK, a transmembrane protein, was needed for the replication of HRV, influenza A virus, and dengue virus. RNASEK localizes to the cell surface and endosomal pathway and closely associates with the vacuolar ATPase (V-ATPase) proton pump. RNASEK is required for endocytosis, and its depletion produces enlarged clathrin-coated pits (CCPs) at the cell surface. These enlarged CCPs contain endocytic cargo and are bound by the scissioning GTPase, DNM2. Loss of RNASEK alters the localization of multiple V-ATPase subunits and lowers the levels of the ATP6AP1 subunit. Together, our results show that RNASEK closely associates with the V-ATPase and is required for its function; its loss prevents the early events of endocytosis and the replication of multiple pathogenic viruses.


Asunto(s)
Virus del Dengue/fisiología , Endorribonucleasas/metabolismo , Virus de la Influenza A/fisiología , Rhinovirus/fisiología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Replicación Viral/fisiología , Endocitosis/fisiología , Endorribonucleasas/genética , Células HeLa , Humanos , ATPasas de Translocación de Protón Vacuolares/genética
19.
Immunity ; 41(6): 1001-12, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25526311

RESUMEN

Decreased HIV-specific CD8(+) T cell proliferation is a hallmark of chronic infection, but the mechanisms of decline are unclear. We analyzed gene expression profiles from antigen-stimulated HIV-specific CD8(+) T cells from patients with controlled and uncontrolled infection and identified caspase-8 as a correlate of dysfunctional CD8(+) T cell proliferation. Caspase-8 activity was upregulated in HIV-specific CD8(+) T cells from progressors and correlated positively with disease progression and programmed cell death-1 (PD-1) expression, but negatively with proliferation. In addition, progressor cells displayed a decreased ability to upregulate membrane-associated caspase-8 activity and increased necrotic cell death following antigenic stimulation, implicating the programmed cell death pathway necroptosis. In vitro necroptosis blockade rescued HIV-specific CD8(+) T cell proliferation in progressors, as did silencing of necroptosis mediator RIPK3. Thus, chronic stimulation leading to upregulated caspase-8 activity contributes to dysfunctional HIV-specific CD8(+) T cell proliferation through activation of necroptosis and increased cell death.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Caspasa 8/metabolismo , Infecciones por VIH/inmunología , VIH/fisiología , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T CD8-positivos/virología , Proliferación Celular/genética , Células Cultivadas , Progresión de la Enfermedad , Activación Enzimática , Regulación de la Expresión Génica , Proteína p24 del Núcleo del VIH/inmunología , Humanos , Necrosis , Fragmentos de Péptidos/inmunología , Receptor de Muerte Celular Programada 1/genética , ARN Interferente Pequeño/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transcriptoma , Carga Viral
20.
Cell Rep ; 9(2): 752-66, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25373910

RESUMEN

RNAi screens have implicated hundreds of host proteins as HIV-1 dependency factors (HDFs). While informative, these early studies overlap poorly due to false positives and false negatives. To ameliorate these issues, we combined information from the existing HDF screens together with new screens performed with multiple orthologous RNAi reagents (MORR). In addition to being traditionally validated, the MORR screens and the historical HDF screens were quantitatively integrated by the adaptation of an established analysis program, RIGER, for the collective interpretation of each gene's phenotypic significance. False positives were addressed by the removal of poorly expressed candidates through gene expression filtering, as well as with GESS, which identifies off-target effects. This workflow produced a quantitatively integrated network of genes that modulate HIV-1 replication. We further investigated the roles of GOLGI49, SEC13, and COG in HIV-1 replication. Collectively, the MORR-RIGER method minimized the caveats of RNAi screening and improved our understanding of HIV-1-host cell interactions.


Asunto(s)
VIH-1/fisiología , Ensayos Analíticos de Alto Rendimiento/métodos , Interacciones Huésped-Patógeno , Interferencia de ARN , Replicación Viral , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Algoritmos , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN , Células HEK293 , Células HeLa , Humanos , Células Jurkat , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...