Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Nat Commun ; 15(1): 4153, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38755212

RESUMO

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.


Assuntos
Proteínas ADAM , Infecções por Cardiovirus , Vírus da Encefalomiocardite , Imunidade Inata , Interferon Tipo I , Helicase IFIH1 Induzida por Interferon , Proteínas de Membrana , Camundongos Knockout , Miocardite , Animais , Vírus da Encefalomiocardite/imunologia , Helicase IFIH1 Induzida por Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/imunologia , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/virologia , Proteínas ADAM/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/imunologia , Camundongos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Miocardite/imunologia , Miocardite/virologia , Humanos , Camundongos Endogâmicos C57BL , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Transdução de Sinais/imunologia , Masculino , Células HEK293
2.
Methods Mol Biol ; 2142: 215-234, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32367370

RESUMO

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.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Testes Genéticos/métodos , Ensaios de Triagem em Larga Escala/métodos , Infecção por Zika virus/diagnóstico , Zika virus/genética , Zika virus/patogenicidade , Células A549 , Proteína 9 Associada à CRISPR/metabolismo , Técnicas de Cultura de Células/métodos , Citometria de Fluxo , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Fenótipo , RNA Guia de Cinetoplastídeos/genética , Carga Viral/métodos , Fatores de Virulência/genética , Fatores de Virulência/isolamento & purificação , Cultura de Vírus/métodos , Zika virus/fisiologia , Infecção por Zika virus/genética , Infecção por Zika virus/patologia
3.
Microb Cell ; 7(5): 129-138, 2020 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-32391394

RESUMO

Sulforaphane (SFN) is a compound [1-isothiocyanato-4-(methylsulfinyl)-butane] found in broccoli and other cruciferous vegetables that is currently of interest because of its potential as a chemopreventive and a chemotherapeutic drug. Recent studies in a diverse range of cellular and animal models have shown that SFN is involved in multiple intracellular pathways that regulate xenobiotic metabolism, inflammation, cell death, cell cycle progression, and epigenetic regulation. In order to better understand the mechanisms of action behind SFN-induced cell death, we undertook an unbiased genome wide screen with the yeast knockout (YKO) library to identify SFN sensitive (SFNS) mutants. The mutants were enriched with knockouts in genes linked to vacuolar function suggesting a link between this organelle and SFN's mechanism of action in yeast. Our subsequent work revealed that SFN increases the vacuolar pH of yeast cells and that varying the vacuolar pH can alter the sensitivity of yeast cells to the drug. In fact, several mutations that lower the vacuolar pH in yeast actually made the cells resistant to SFN (SFNR). Finally, we show that human lung cancer cells with more acidic compartments are also SFNR suggesting that SFN's mechanism of action identified in yeast may carry over to higher eukaryotic cells.

4.
J Immunol ; 203(7): 1999-2010, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31471524

RESUMO

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.


Assuntos
Apresentação de Antígeno , Antígeno B7-H1/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Fator Regulador 2 de Interferon/deficiência , Proteínas de Neoplasias/imunologia , Neoplasias , Evasão Tumoral , Antígeno B7-H1/genética , Linfócitos T CD8-Positivos/patologia , Regulação para Baixo/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Células HEK293 , Células HeLa , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Fator Regulador 2 de Interferon/imunologia , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia
5.
Nat Med ; 25(9): 1377-1384, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31501601

RESUMO

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.


Assuntos
Fármacos Anti-HIV/farmacologia , Proteínas do Capsídeo/antagonistas & inibidores , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Indazóis/farmacologia , Piridinas/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Fármacos Anti-HIV/uso terapêutico , Capsídeo/efeitos dos fármacos , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , DNA Viral/efeitos dos fármacos , Preparações de Ação Retardada , Farmacorresistência Viral/efeitos dos fármacos , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/genética , HIV-1/patogenicidade , HIV-2/efeitos dos fármacos , HIV-2/patogenicidade , Humanos , Indazóis/uso terapêutico , Adesão à Medicação , Camundongos , Piridinas/uso terapêutico
6.
J Life Sci (Westlake Village) ; 1(1): 39-45, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31448372

RESUMO

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.

7.
Proc Natl Acad Sci U S A ; 116(14): 7043-7052, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30894498

RESUMO

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.


Assuntos
Citomegalovirus/fisiologia , Células Epiteliais/metabolismo , Complexos Multiproteicos/metabolismo , Transdução de Sinais , Proteínas Virais/metabolismo , Tropismo Viral/fisiologia , Células A549 , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Epiteliais/patologia , Células Epiteliais/virologia , Células HEK293 , Células HeLa , Humanos , Complexos Multiproteicos/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Virais/genética
8.
mBio ; 10(1)2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30723129

RESUMO

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.


Assuntos
Proteínas ADAM/metabolismo , Infecções por Cardiovirus/genética , Resistência à Doença , Vírus da Encefalomiocardite/crescimento & desenvolvimento , Proteínas de Membrana/metabolismo , Animais , Linhagem Celular , Técnicas de Inativação de Genes , Testes Genéticos , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos
9.
iScience ; 12: 270-279, 2019 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-30716700

RESUMO

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.

10.
PLoS Pathog ; 15(1): e1007532, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30640957

RESUMO

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.


Assuntos
Endossomos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/fisiologia , Células A549/metabolismo , Animais , Antivirais/metabolismo , Células COS/metabolismo , Chlorocebus aethiops , Endossomos/virologia , Células HEK293/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A/patogenicidade , Interferons/metabolismo , Vírus Lassa/patogenicidade , Imagem Óptica/métodos , Transporte Proteico , Internalização do Vírus
11.
J Virol ; 93(2)2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30355696

RESUMO

The interferon-induced transmembrane proteins (IFITMs) are a family of highly related antiviral factors that affect numerous viruses at two steps: in target cells by sequestering incoming viruses in endosomes and in producing cells by leading to the production of virions that package IFITMs and exhibit decreased infectivity. While most studies have focused on the former, little is known about the regulation of the negative imprinting of virion particle infectivity by IFITMs and about its relationship with target cell protection. Using a panel of IFITM3 mutants against HIV-1, we have explored these issues as well as others related to the biology of IFITM3, in particular virion packaging, stability, the relation to CD63/multivesicular bodies (MVBs), the modulation of cholesterol levels, and the relationship between negative imprinting of virions and target cell protection. The results that we have obtained exclude a role for cholesterol and indicate that CD63 accumulation does not directly relate to an antiviral behavior. We have defined regions that modulate the two antiviral properties of IFITM3 as well as novel domains that modulate protein stability and that, in so doing, influence the extent of its packaging into virions. The results that we have obtained, however, indicate that, even in the context of an IFITM-susceptible virus, IFITM3 packaging is not sufficient for negative imprinting. Finally, while most mutations concomitantly affect target cell protection and negative imprinting, a region in the C-terminal domain (CTD) exhibits a differential behavior, potentially highlighting the regulatory role that this domain may play in the two antiviral activities of IFITM3.IMPORTANCE IFITM proteins have been associated with the sequestration of incoming virions in endosomes (target cell protection) and with the production of virion particles that incorporate IFITMs and exhibit decreased infectivity (negative imprinting of virion infectivity). How the latter is regulated and whether these two antiviral properties are related remain unknown. By examining the behavior of a large panel of IFITM3 mutants against HIV-1, we determined that IFITM3 mutants are essentially packaged into virions proportionally to their intracellular levels of expression. However, even in the context of an IFITM-susceptible virus, IFITM3 packaging is not sufficient for the antiviral effects. Most mutations were found to concomitantly affect both antiviral properties of IFITM3, but one CTD mutant exhibited a divergent behavior, possibly highlighting a novel regulatory role for this domain. These findings thus advance our comprehension of how this class of broad antiviral restriction factors acts.


Assuntos
HIV-1/fisiologia , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Mutação , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Vírion/fisiologia , Colesterol/metabolismo , Endossomos , Células HEK293 , Células HeLa , Humanos , Proteínas de Membrana/genética , Domínios Proteicos , Estabilidade Proteica , Proteínas de Ligação a RNA/genética , Tetraspanina 30/metabolismo , Montagem de Vírus
12.
Cell Host Microbe ; 24(3): 392-404.e8, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30173955

RESUMO

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.


Assuntos
Capsídeo/metabolismo , Núcleo Celular/virologia , DNA Viral/genética , Infecções por HIV/metabolismo , HIV-1/fisiologia , Integração Viral , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , DNA Viral/metabolismo , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/genética , Interações Hospedeiro-Patógeno , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ligação Proteica , Replicação Viral , Fatores de Poliadenilação e Clivagem de mRNA/genética
13.
PLoS Comput Biol ; 14(4): e1006093, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29677181

RESUMO

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.


Assuntos
Antígenos HIV/química , Antígenos HIV/imunologia , Proteína gp120 do Envelope de HIV/química , Proteína gp120 do Envelope de HIV/imunologia , Polissacarídeos/química , Polissacarídeos/imunologia , Algoritmos , Sequência de Aminoácidos , Anticorpos Neutralizantes , Teorema de Bayes , Sítios de Ligação , Biologia Computacional , Epitopos/química , Epitopos/genética , Glicosilação , HIV/química , HIV/imunologia , Anticorpos Anti-HIV , Antígenos HIV/genética , Proteína gp120 do Envelope de HIV/genética , Humanos , Aprendizado de Máquina , Modelos Moleculares , Engenharia de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Biologia de Sistemas
14.
Curr Opin Virol ; 29: 87-100, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29684735

RESUMO

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.


Assuntos
Sistemas CRISPR-Cas , Testes Genéticos , Interações Hospedeiro-Patógeno , Viroses/virologia , Vírus/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Flavivirus/genética , Flavivirus/metabolismo , Humanos , Receptores Virais/metabolismo , Fluxo de Trabalho
15.
Nature ; 538(7625): 350-355, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27680706

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Colo/metabolismo , Epitélio/metabolismo , Receptores Frizzled/metabolismo , Animais , Antígenos/metabolismo , Proteínas de Bactérias/química , Toxinas Bacterianas/química , Sítios de Ligação , Células CHO , Sistemas CRISPR-Cas , Linhagem Celular , Clostridioides difficile/patogenicidade , Cricetulus , Feminino , Receptores Frizzled/química , Receptores Frizzled/deficiência , Receptores Frizzled/genética , Técnicas de Inativação de Genes , Humanos , Masculino , Camundongos , Camundongos Knockout , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Organoides/metabolismo , Domínios Proteicos , Proteoglicanas/metabolismo , Fatores de Virulência/metabolismo , Proteínas Wnt/metabolismo
16.
Cell Rep ; 15(11): 2323-30, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27268505

RESUMO

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.


Assuntos
Antígenos de Diferenciação/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA/metabolismo , Replicação Viral/fisiologia , Zika virus/fisiologia , Células A549 , Animais , Efeito Citopatogênico Viral , Deleção de Genes , Loci Gênicos , Células HeLa , Humanos , Camundongos , Transporte Proteico , Infecção por Zika virus
17.
Cell Rep ; 16(1): 232-246, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27342126

RESUMO

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.


Assuntos
Vírus da Dengue/genética , Genômica/métodos , Zika virus/genética , Sistemas CRISPR-Cas , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Testes Genéticos , Células HeLa , Interações Hospedeiro-Patógeno/genética , Humanos , Membranas Intracelulares/metabolismo , Ligação Proteica , Mapas de Interação de Proteínas , Interferência de RNA , Replicação Viral
18.
Adv Virus Res ; 94: 1-51, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26997589

RESUMO

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.


Assuntos
Sistemas CRISPR-Cas , Genômica/métodos , Haploidia , Interações Hospedeiro-Patógeno/genética , Interferência de RNA , Vírus/patogenicidade , Proteínas de Bactérias , Proteína 9 Associada à CRISPR , Endonucleases , Técnicas de Inativação de Genes , Testes Genéticos/métodos , Humanos/virologia , Mutagênese Insercional , RNA Interferente Pequeno/genética
19.
Cell Host Microbe ; 18(6): 723-35, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26651948

RESUMO

Several systems-level datasets designed to dissect host-pathogen interactions during influenza A infection have been reported. However, apparent discordance among these data has hampered their full utility toward advancing mechanistic and therapeutic knowledge. To collectively reconcile these datasets, we performed a meta-analysis of data from eight published RNAi screens and integrated these data with three protein interaction datasets, including one generated within the context of this study. Further integration of these data with global virus-host interaction analyses revealed a functionally validated biochemical landscape of the influenza-host interface, which can be queried through a simplified and customizable web portal (http://www.metascape.org/IAV). Follow-up studies revealed that the putative ubiquitin ligase UBR4 associates with the viral M2 protein and promotes apical transport of viral proteins. Taken together, the integrative analysis of influenza OMICs datasets illuminates a viral-host network of high-confidence human proteins that are essential for influenza A virus replication.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Proteínas do Citoesqueleto/metabolismo , Interações Hospedeiro-Patógeno , Vírus da Influenza A/fisiologia , Proteínas da Matriz Viral/metabolismo , Liberação de Vírus , Animais , Linhagem Celular , Biologia Computacional , Citometria de Fluxo , Humanos , Imunoprecipitação , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Ligação Proteica , Mapas de Interação de Proteínas , Transporte Proteico , Ubiquitina-Proteína Ligases
20.
Cell Rep ; 13(8): 1717-31, 2015 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-26586435

RESUMO

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.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , HIV-1/genética , Integração Viral/genética , Replicação Viral/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Citosol/metabolismo , Replicação do DNA/genética , Genoma Viral/genética , Infecções por HIV/genética , Infecções por HIV/metabolismo , Células HeLa , Humanos , Transcrição Reversa/genética , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA