Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nature ; 543(7647): 723-727, 2017 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-28329770

RESUMEN

Cancer somatic mutations can generate neoantigens that distinguish malignant from normal cells. However, the personalized identification and validation of neoantigens remains a major challenge. Here we discover neoantigens in human mantle-cell lymphomas by using an integrated genomic and proteomic strategy that interrogates tumour antigen peptides presented by major histocompatibility complex (MHC) class I and class II molecules. We applied this approach to systematically characterize MHC ligands from 17 patients. Remarkably, all discovered neoantigenic peptides were exclusively derived from the lymphoma immunoglobulin heavy- or light-chain variable regions. Although we identified MHC presentation of private polymorphic germline alleles, no mutated peptides were recovered from non-immunoglobulin somatically mutated genes. Somatic mutations within the immunoglobulin variable region were almost exclusively presented by MHC class II. We isolated circulating CD4+ T cells specific for immunoglobulin-derived neoantigens and found these cells could mediate killing of autologous lymphoma cells. These results demonstrate that an integrative approach combining MHC isolation, peptide identification, and exome sequencing is an effective platform to uncover tumour neoantigens. Application of this strategy to human lymphoma implicates immunoglobulin neoantigens as targets for lymphoma immunotherapy.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Región Variable de Inmunoglobulina/inmunología , Linfoma de Células del Manto/inmunología , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/inmunología , Citotoxicidad Inmunológica , Análisis Mutacional de ADN , Epítopos de Linfocito T/inmunología , Exoma/genética , Genómica , Antígenos HLA-D/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Región Variable de Inmunoglobulina/química , Región Variable de Inmunoglobulina/genética , Inmunoterapia/tendencias , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/patología , Linfoma de Células del Manto/terapia , Mutación , Proteómica
2.
Nature ; 535(7610): 159-63, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27383987

RESUMEN

The Flaviviridae are a family of viruses that cause severe human diseases. For example, dengue virus (DENV) is a rapidly emerging pathogen causing an estimated 100 million symptomatic infections annually worldwide. No approved antivirals are available to date and clinical trials with a tetravalent dengue vaccine showed disappointingly low protection rates. Hepatitis C virus (HCV) also remains a major medical problem, with 160 million chronically infected patients worldwide and only expensive treatments available. Despite distinct differences in their pathogenesis and modes of transmission, the two viruses share common replication strategies. A detailed understanding of the host functions that determine viral infection is lacking. Here we use a pooled CRISPR genetic screening strategy to comprehensively dissect host factors required for these two highly important Flaviviridae members. For DENV, we identified endoplasmic-reticulum (ER)-associated multi-protein complexes involved in signal sequence recognition, N-linked glycosylation and ER-associated degradation. DENV replication was nearly completely abrogated in cells deficient in the oligosaccharyltransferase (OST) complex. Mechanistic studies pinpointed viral RNA replication and not entry or translation as the crucial step requiring the OST complex. Moreover, we show that viral non-structural proteins bind to the OST complex. The identified ER-associated protein complexes were also important for infection by other mosquito-borne flaviviruses including Zika virus, an emerging pathogen causing severe birth defects. By contrast, the most significant genes identified in the HCV screen were distinct and included viral receptors, RNA-binding proteins and enzymes involved in metabolism. We found an unexpected link between intracellular flavin adenine dinucleotide (FAD) levels and HCV replication. This study shows notable divergence in host-depenency factors between DENV and HCV, and illuminates new host targets for antiviral therapy.


Asunto(s)
Sistemas CRISPR-Cas/genética , Virus del Dengue/fisiología , Genoma Humano/genética , Hepacivirus/fisiología , Factores Celulares Derivados del Huésped/genética , Interacciones Huésped-Patógeno/genética , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Descubrimiento de Drogas , Retículo Endoplásmico/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Flavina-Adenina Dinucleótido/biosíntesis , Flavina-Adenina Dinucleótido/metabolismo , Infecciones por Flavivirus/genética , Infecciones por Flavivirus/virología , Glicosilación , Hexosiltransferasas/deficiencia , Hexosiltransferasas/genética , Hexosiltransferasas/metabolismo , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Terapia Molecular Dirigida , Unión Proteica , Señales de Clasificación de Proteína , Proteínas de Unión al ARN/genética , Receptores Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Virus Zika/metabolismo
3.
Proc Natl Acad Sci U S A ; 112(2): 319-25, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25516984

RESUMEN

Translation initiation can occur by multiple pathways. To delineate these pathways by single-molecule methods, fluorescently labeled ribosomal subunits are required. Here, we labeled human 40S ribosomal subunits with a fluorescent SNAP-tag at ribosomal protein eS25 (RPS25). The resulting ribosomal subunits could be specifically labeled in living cells and in vitro. Using single-molecule Förster resonance energy transfer (FRET) between RPS25 and domain II of the hepatitis C virus (HCV) internal ribosome entry site (IRES), we measured the rates of 40S subunit arrival to the HCV IRES. Our data support a single-step model of HCV IRES recruitment to 40S subunits, irreversible on the initiation time scale. We furthermore demonstrated that after binding, the 40S:HCV IRES complex is conformationally dynamic, undergoing slow large-scale rearrangements. Addition of translation extracts suppresses these fluctuations, funneling the complex into a single conformation on the 80S assembly pathway. These findings show that 40S:HCV IRES complex formation is accompanied by dynamic conformational rearrangements that may be modulated by initiation factors.


Asunto(s)
Hepacivirus/genética , Hepacivirus/metabolismo , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/genética , Sistemas CRISPR-Cas , Transferencia Resonante de Energía de Fluorescencia , Técnicas de Inactivación de Genes , Células HeLa , Hepacivirus/patogenicidad , Interacciones Huésped-Patógeno , Humanos , Cinética , Datos de Secuencia Molecular , O(6)-Metilguanina-ADN Metiltransferasa/genética , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Iniciación de la Cadena Peptídica Traduccional , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Ribosómicas/antagonistas & inhibidores , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo
4.
Proc Natl Acad Sci U S A ; 112(46): 14337-42, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26489655

RESUMEN

Staphylococcus aureus is both a transient skin colonizer and a formidable human pathogen, ranking among the leading causes of skin and soft tissue infections as well as severe pneumonia. The secreted bacterial α-toxin is essential for S. aureus virulence in these epithelial diseases. To discover host cellular factors required for α-toxin cytotoxicity, we conducted a genetic screen using mutagenized haploid human cells. Our screen identified a cytoplasmic member of the adherens junctions, plekstrin-homology domain containing protein 7 (PLEKHA7), as the second most significantly enriched gene after the known α-toxin receptor, a disintegrin and metalloprotease 10 (ADAM10). Here we report a new, unexpected role for PLEKHA7 and several components of cellular adherens junctions in controlling susceptibility to S. aureus α-toxin. We find that despite being injured by α-toxin pore formation, PLEKHA7 knockout cells recover after intoxication. By infecting PLEKHA7(-/-) mice with methicillin-resistant S. aureus USA300 LAC strain, we demonstrate that this junctional protein controls disease severity in both skin infection and lethal S. aureus pneumonia. Our results suggest that adherens junctions actively control cellular responses to a potent pore-forming bacterial toxin and identify PLEKHA7 as a potential nonessential host target to reduce S. aureus virulence during epithelial infections.


Asunto(s)
Uniones Adherentes/metabolismo , Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Staphylococcus aureus Resistente a Meticilina/metabolismo , Infecciones Estafilocócicas/metabolismo , Vasculitis/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Uniones Adherentes/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Toxinas Bacterianas/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Proteínas Hemolisinas/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones , Ratones Noqueados , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/patología , Vasculitis/genética , Vasculitis/microbiología , Vasculitis/patología
5.
Circ Res ; 115(6): 556-66, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25015077

RESUMEN

RATIONALE: Viral myocarditis is a life-threatening illness that may lead to heart failure or cardiac arrhythmias. A major causative agent for viral myocarditis is the B3 strain of coxsackievirus, a positive-sense RNA enterovirus. However, human cardiac tissues are difficult to procure in sufficient enough quantities for studying the mechanisms of cardiac-specific viral infection. OBJECTIVE: This study examined whether human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) could be used to model the pathogenic processes of coxsackievirus-induced viral myocarditis and to screen antiviral therapeutics for efficacy. METHODS AND RESULTS: hiPSC-CMs were infected with a luciferase-expressing coxsackievirus B3 strain (CVB3-Luc). Brightfield microscopy, immunofluorescence, and calcium imaging were used to characterize virally infected hiPSC-CMs for alterations in cellular morphology and calcium handling. Viral proliferation in hiPSC-CMs was quantified using bioluminescence imaging. Antiviral compounds including interferonß1, ribavirin, pyrrolidine dithiocarbamate, and fluoxetine were tested for their capacity to abrogate CVB3-Luc proliferation in hiPSC-CMs in vitro. The ability of these compounds to reduce CVB3-Luc proliferation in hiPSC-CMs was consistent with reported drug effects in previous studies. Mechanistic analyses via gene expression profiling of hiPSC-CMs infected with CVB3-Luc revealed an activation of viral RNA and protein clearance pathways after interferonß1 treatment. CONCLUSIONS: This study demonstrates that hiPSC-CMs express the coxsackievirus and adenovirus receptor, are susceptible to coxsackievirus infection, and can be used to predict antiviral drug efficacy. Our results suggest that the hiPSC-CM/CVB3-Luc assay is a sensitive platform that can screen novel antiviral therapeutics for their effectiveness in a high-throughput fashion.


Asunto(s)
Antivirales/uso terapéutico , Enterovirus Humano B/aislamiento & purificación , Infecciones por Enterovirus/tratamiento farmacológico , Modelos Cardiovasculares , Miocarditis/tratamiento farmacológico , Miocitos Cardíacos/patología , Células Madre Pluripotentes/patología , Antivirales/farmacología , Calcio/metabolismo , Proliferación Celular , Células Cultivadas , Evaluación Preclínica de Medicamentos , Infecciones por Enterovirus/metabolismo , Humanos , Técnicas In Vitro , Miocarditis/metabolismo , Miocarditis/virología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/virología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/virología , ARN Viral/metabolismo , Resultado del Tratamiento
6.
J Infect Dis ; 212 Suppl 2: S410-3, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25999057

RESUMEN

As of 25 March 2015, the largest recorded outbreak of Ebola virus infection is ongoing, with almost 25 000 cases and >10 000 deaths. There are 5 genetically and antigenically distinct species within the genus Ebolavirus. Limited cross-reactivity and protection is observed between these 5 Ebolavirus species, which complicates vaccine development. However, on the basis of sequence homology between the 5 Ebolavirus species, we hypothesize that conserved epitopes are present on the viral glycoprotein (GP), which can be targeted by antibodies. In the current study, a panel of mouse monoclonal antibodies was isolated and characterized using an enzyme-linked immunosorbent assay (ELISA) to determine cross-reactivity, avidity, and competition for epitope binding; Western blot analysis was also performed. Four monoclonal antibodies were identified by ELISA as cross-reacting with the GPs of all 5 Ebolavirus species. The identification of cross-reactive antibodies that bind the GPs of all known Ebolavirus species will give us important insight into the presence of conserved epitopes on the viral GP. These data will be crucial for the development of novel therapeutics and diagnostic assays.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Reacciones Cruzadas/inmunología , Ebolavirus/inmunología , Animales , Antígenos Virales/inmunología , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática/métodos , Epítopos/inmunología , Glicoproteínas/inmunología , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/virología , Hibridomas/inmunología , Hibridomas/virología , Ratones , Proteínas Virales/inmunología
7.
bioRxiv ; 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38712102

RESUMEN

Dengue virus (DENV) can hijack non-neutralizing IgG antibodies to facilitate its uptake into target cells expressing Fc gamma receptors (FcgR) - a process known as antibody-dependent enhancement (ADE) of infection. Beyond a requirement for FcgR, host dependency factors for this non-canonical infection route remain unknown. To identify cellular factors exclusively required for ADE, here, we performed CRISPR knockout screens in an in vitro system permissive to infection only in the presence of IgG antibodies. Validating our approach, a top hit was FcgRIIa, which facilitates binding and internalization of IgG-bound DENV but is not required for canonical infection. Additionally, we identified host factors with no previously described role in DENV infection, including TBC1D24 and SV2B, both of which have known functions in regulated secretion. Using genetic knockout and trans-complemented cells, we validated a functional requirement for these host factors in ADE assays performed with monoclonal antibodies and polyclonal sera in multiple cell lines and using all four DENV serotypes. We show that knockout of TBC1D24 or SV2B impaired binding of IgG-DENV complexes to cells without affecting FcgRIIa expression levels. Thus, we identify cellular factors beyond FcgR that are required for ADE of DENV infection. Our findings represent a first step towards advancing fundamental knowledge behind the biology of ADE that can ultimately be exploited to inform vaccination and therapeutic approaches.

8.
JHEP Rep ; 5(11): 100893, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37929228

RESUMEN

Background & Aims: Bulevirtide (BLV) is a small lipopeptide agent that specifically binds to the sodium taurocholate cotransporting polypeptide (NTCP) bile salt transporter and HBV/HDV receptor on the surface of human hepatocytes and inhibits HDV and HBV entry. As a satellite virus of HBV, HDV virions are formed after assembly of HDV RNA with the HBV envelope proteins (HBsAg). Because both viruses exist as eight different genotypes, this creates a potential for high diversity in the HBV/HDV combinations. To investigate the sensitivity of various combinations of HBV/HDV genotypes to BLV, clinical and laboratory strains were assessed. Methods: For the laboratory strains, the different envelopes from HBV genotypes A through H were combined with HDV genotypes 1-8 in cotransfection assays. Clinical plasma isolates were obtained from clinical studies and academic collaborations to maximise the diversity of HBV/HDV genotypes tested. Results: The mean BLV EC50 against HDV laboratory strains ranged from 0.44 to 0.64 nM. Regardless of HBV and HDV genotypes, the clinical isolates showed similar sensitivities to BLV with mean values that ranged from 0.2 to 0.73 nM. Conclusions: These data support the use of BLV in patients infected with any HBV/HDV genotypes. Impact and implications: This study describes the potent activity of BLV against multiple laboratory strains spanning all HBV/HDV A-H/1-8 genotype combinations and the most diverse collection of HDV clinical samples tested to date, including HBV/HDV genotype combinations less frequently observed in the clinic. Overall, all isolates and laboratory strains displayed similar in vitro nanomolar sensitivity to BLV. This broad-spectrum antiviral activity of BLV has direct implications on potential simplified treatment for any patient infected with HDV, regardless of genotype, and supports the new 2023 EASL Clinical Practice Guidelines on HDV that recommend antiviral treatment for all patients with CHD.

9.
mBio ; 10(4)2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31409686

RESUMEN

Encephalomyocarditis virus (EMCV) is an animal pathogen and an important model organism, whose receptor requirements are poorly understood. Here, we employed a genome-wide haploid genetic screen to identify novel EMCV host factors. In addition to the previously described picornavirus receptors sialic acid and glycosaminoglycans, this screen unveiled important new host factors for EMCV. These factors include components of the fibroblast growth factor (FGF) signaling pathway, such as the potential receptors FGFR1 and ADAM9, a cell-surface metalloproteinase. By employing various knockout cells, we confirmed the importance of the identified host factors for EMCV infection. The largest reduction in infection efficiency was observed in cells lacking ADAM9. Pharmacological inhibition of the metalloproteinase activity of ADAM9 did not affect virus infection. Moreover, reconstitution of inactive ADAM9 in knockout cells restored susceptibility to EMCV, pointing to a proteinase-independent role of ADAM9 in mediating EMCV infection. Using neutralization assays with ADAM9-specific antiserum and soluble receptor proteins, we provided evidence for a role of ADAM9 in EMCV entry. Finally, binding assays showed that ADAM9 facilitates attachment of EMCV to the cell surface. Together, our findings reveal a role for ADAM9 as a novel receptor or cofactor for EMCV.IMPORTANCE EMCV is an animal pathogen that causes acute viral infections, usually myocarditis or encephalitis. It is thought to circulate mainly among rodents, from which it is occasionally transmitted to other animal species, including humans. EMCV causes fatal outbreaks of myocarditis and encephalitis in pig farms and zoos, making it an important veterinary pathogen. Although EMCV has been widely used as a model to study mechanisms of viral disease in mice, little is known about its entry mechanism. Here, we employ a haploid genetic screen for EMCV host factors and identify an essential role for ADAM9 in EMCV entry.


Asunto(s)
Proteínas ADAM/metabolismo , Infecciones por Cardiovirus/virología , Virus de la Encefalomiocarditis/fisiología , Proteínas de la Membrana/metabolismo , Internalización del Virus , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/genética , Animales , Infecciones por Cardiovirus/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Virus de la Encefalomiocarditis/metabolismo , Técnicas de Inactivación de Genes , Genoma Humano/genética , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Acoplamiento Viral , Replicación Viral
10.
Nat Microbiol ; 4(12): 2369-2382, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31384002

RESUMEN

Flaviviruses, including dengue virus (DENV) and Zika virus (ZIKV), cause severe human disease. Co-opting cellular factors for viral translation and viral genome replication at the endoplasmic reticulum is a shared replication strategy, despite different clinical outcomes. Although the protein products of these viruses have been studied in depth, how the RNA genomes operate inside human cells is poorly understood. Using comprehensive identification of RNA-binding proteins by mass spectrometry (ChIRP-MS), we took an RNA-centric viewpoint of flaviviral infection and identified several hundred proteins associated with both DENV and ZIKV genomic RNA in human cells. Genome-scale knockout screens assigned putative functional relevance to the RNA-protein interactions observed by ChIRP-MS. The endoplasmic-reticulum-localized RNA-binding proteins vigilin and ribosome-binding protein 1 directly bound viral RNA and each acted at distinct stages in the life cycle of flaviviruses. Thus, this versatile strategy can elucidate features of human biology that control the pathogenesis of clinically relevant viruses.


Asunto(s)
Infecciones por Flavivirus/virología , Flavivirus/genética , Flavivirus/fisiología , ARN Viral/genética , Sistemas CRISPR-Cas , Proteínas Portadoras , Línea Celular , Virus del Dengue/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Flavivirus/patogenicidad , Técnicas de Inactivación de Genes , Interacciones Huésped-Patógeno/genética , Humanos , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Replicación Viral , Virus Zika/genética
11.
Cell Chem Biol ; 25(10): 1231-1241.e4, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30078634

RESUMEN

The oligosaccharyltransferase (OST) is a multisubunit enzyme complex that N-glycosylates proteins in the secretory pathway and is considered to be constitutive and unregulated. However, small-molecule OST inhibitors such as NGI-1 provide a pharmacological approach for regulating N-linked glycosylation. Herein we design cell models with knockout of each OST catalytic subunit (STT3A or STT3B) to screen the activity of NGI-1 and its analogs. We show that NGI-1 targets the function of both STT3A and STT3B and use structure-activity relationships to guide synthesis of catalytic subunit-specific inhibitors. Using this approach, pharmacophores that increase STT3B selectivity are characterized and an STT3B-specific inhibitor is identified. This inhibitor has discrete biological effects on endogenous STT3B target proteins such as COX2 but does not activate the cellular unfolded protein response. Together this work demonstrates that subsets of glycoproteins can be regulated through pharmacologic inhibition of N-linked glycosylation.


Asunto(s)
Benzamidas/química , Benzamidas/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Hexosiltransferasas/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Sulfonamidas/química , Sulfonamidas/farmacología , Dominio Catalítico , Evaluación Preclínica de Medicamentos , Técnicas de Inactivación de Genes , Células HEK293 , Hexosiltransferasas/genética , Hexosiltransferasas/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Relación Estructura-Actividad
12.
Cell Rep ; 21(11): 3032-3039, 2017 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-29241533

RESUMEN

The mosquito-borne flaviviruses include important human pathogens such as dengue, Zika, West Nile, and yellow fever viruses, which pose a serious threat for global health. Recent genetic screens identified endoplasmic reticulum (ER)-membrane multiprotein complexes, including the oligosaccharyltransferase (OST) complex, as critical flavivirus host factors. Here, we show that a chemical modulator of the OST complex termed NGI-1 has promising antiviral activity against flavivirus infections. We demonstrate that NGI-1 blocks viral RNA replication and that antiviral activity does not depend on inhibition of the N-glycosylation function of the OST. Viral mutants adapted to replicate in cells deficient of the OST complex showed resistance to NGI-1 treatment, reinforcing the on-target activity of NGI-1. Lastly, we show that NGI-1 also has strong antiviral activity in primary and disease-relevant cell types. This study provides an example for advancing from the identification of genetic determinants of infection to a host-directed antiviral compound with broad activity against flaviviruses.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , Virus del Dengue/efectos de los fármacos , Hexosiltransferasas/genética , Interacciones Huésped-Patógeno/efectos de los fármacos , Proteínas de la Membrana/genética , Sulfonamidas/farmacología , Replicación Viral/efectos de los fármacos , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Células HEK293 , Hexosiltransferasas/antagonistas & inhibidores , Hexosiltransferasas/deficiencia , Humanos , Luciferasas , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/deficiencia , Pruebas de Sensibilidad Microbiana , Transducción de Señal , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/crecimiento & desarrollo , Virus de la Fiebre Amarilla/efectos de los fármacos , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/crecimiento & desarrollo , Virus Zika/efectos de los fármacos , Virus Zika/genética , Virus Zika/crecimiento & desarrollo
13.
Cancer Res ; 76(19): 5810-5821, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27503929

RESUMEN

Anthracyclines are among the most effective yet most toxic drugs used in the oncology clinic. The nucleosome-remodeling SWI/SNF complex, a potent tumor suppressor, is thought to promote sensitivity to anthracyclines by recruiting topoisomerase IIa (TOP2A) to DNA and increasing double-strand breaks. In this study, we discovered a novel mechanism through which SWI/SNF influences resistance to the widely used anthracycline doxorubicin based on the use of a forward genetic screen in haploid human cells, followed by a rigorous single and double-mutant epistasis analysis using CRISPR/Cas9-mediated engineering. Doxorubicin resistance conferred by loss of the SMARCB1 subunit of the SWI/SNF complex was caused by transcriptional upregulation of a single gene, encoding the multidrug resistance pump ABCB1. Remarkably, both ABCB1 upregulation and doxorubicin resistance caused by SMARCB1 loss were dependent on the function of SMARCA4, a catalytic subunit of the SWI/SNF complex. We propose that residual SWI/SNF complexes lacking SMARCB1 are vital determinants of drug sensitivity, not just to TOP2A-targeted agents, but to the much broader range of cancer drugs effluxed by ABCB1. Cancer Res; 76(19); 5810-21. ©2016 AACR.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN Helicasas/fisiología , Proteínas Nucleares/fisiología , Proteína SMARCB1/fisiología , Factores de Transcripción/fisiología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Haploidia , Humanos , Transcripción Genética
14.
Elife ; 52016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27996937

RESUMEN

The comprehensive understanding of cellular signaling pathways remains a challenge due to multiple layers of regulation that may become evident only when the pathway is probed at different levels or critical nodes are eliminated. To discover regulatory mechanisms in canonical WNT signaling, we conducted a systematic forward genetic analysis through reporter-based screens in haploid human cells. Comparison of screens for negative, attenuating and positive regulators of WNT signaling, mediators of R-spondin-dependent signaling and suppressors of constitutive signaling induced by loss of the tumor suppressor adenomatous polyposis coli or casein kinase 1α uncovered new regulatory features at most levels of the pathway. These include a requirement for the transcription factor AP-4, a role for the DAX domain of AXIN2 in controlling ß-catenin transcriptional activity, a contribution of glycophosphatidylinositol anchor biosynthesis and glypicans to R-spondin-potentiated WNT signaling, and two different mechanisms that regulate signaling when distinct components of the ß-catenin destruction complex are lost. The conceptual and methodological framework we describe should enable the comprehensive understanding of other signaling systems.


Asunto(s)
Regulación de la Expresión Génica , Redes Reguladoras de Genes , Pruebas Genéticas/métodos , Vía de Señalización Wnt , Quinasa de la Caseína I/deficiencia , Proteínas del Citoesqueleto/deficiencia , Genes Reporteros , Haploidia , Humanos , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
15.
Science ; 350(6264): 1092-6, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26472760

RESUMEN

Although the genes essential for life have been identified in less complex model organisms, their elucidation in human cells has been hindered by technical barriers. We used extensive mutagenesis in haploid human cells to identify approximately 2000 genes required for optimal fitness under culture conditions. To study the principles of genetic interactions in human cells, we created a synthetic lethality network focused on the secretory pathway based exclusively on mutations. This revealed a genetic cross-talk governing Golgi homeostasis, an additional subunit of the human oligosaccharyltransferase complex, and a phosphatidylinositol 4-kinase ß adaptor hijacked by viruses. The synthetic lethality map parallels observations made in yeast and projects a route forward to reveal genetic networks in diverse aspects of human cell biology.


Asunto(s)
Redes Reguladoras de Genes , Genes Esenciales , Genes Letales , Aptitud Genética/genética , Haploidia , Aparato de Golgi/genética , Hexosiltransferasas/genética , Humanos , Proteínas de la Membrana/genética , Mutagénesis Insercional , Mutación , Saccharomyces cerevisiae/genética
16.
Elife ; 42015 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-26565589

RESUMEN

Recognition and elimination of tumor cells by the immune system is crucial for limiting tumor growth. Natural killer (NK) cells become activated when the receptor NKG2D is engaged by ligands that are frequently upregulated in primary tumors and on cancer cell lines. However, the molecular mechanisms driving NKG2D ligand expression on tumor cells are not well defined. Using a forward genetic screen in a tumor-derived human cell line, we identified several novel factors supporting expression of the NKG2D ligand ULBP1. Our results show stepwise contributions of independent pathways working at multiple stages of ULBP1 biogenesis. Deeper investigation of selected hits from the screen showed that the transcription factor ATF4 drives ULBP1 gene expression in cancer cell lines, while the RNA-binding protein RBM4 supports ULBP1 expression by suppressing a novel alternatively spliced isoform of ULBP1 mRNA. These findings offer insight into the stress pathways that alert the immune system to danger.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Factor de Transcripción Activador 4/metabolismo , Línea Celular Tumoral , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Pruebas Genéticas , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Proteínas de Unión al ARN/metabolismo
17.
Front Microbiol ; 3: 295, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22936928

RESUMEN

An emerging model for investigating virus-host interactions in hyperthermophilic Archaea is the Fusellovirus-Sulfolobus system. The host, Sulfolobus, is a hyperthermophilic acidophile endemic to sulfuric hot springs worldwide. The Fuselloviruses, also known as Sulfolobus Spindle-shaped Viruses (SSVs), are "lemon" or "spindle"-shaped double-stranded DNA viruses, which are also found worldwide. Although a few studies have addressed the host-range for the type virus, Sulfolobus Spindle-shaped Virus 1 (SSV1), using common Sulfolobus strains, a comprehensive host-range study for SSV-Sulfolobus systems has not been performed. Herein, we examine six bona fide SSV strains (SSV1, SSV2, SSV3, SSVL1, SSVK1, SSVRH) and their respective infection characteristics on multiple hosts from the family Sulfolobaceae. A spot-on-lawn or "halo" assay was employed to determine SSV infectivity (and host susceptibility) in parallel challenges of multiple SSVs on a lawn of a single Sulfolobus strain. Different SSVs have different host-ranges with SSV1 exhibiting the narrowest host-range and SSVRH exhibiting the broadest host range. In contrast to previous reports, SSVs can infect hosts beyond the genus Sulfolobus. Furthermore, geography does not appear to be a reliable predictor of Sulfolobus susceptibility to infection by any given SSV. The ability for SSVs to infect susceptible Sulfolobus host does not appear to change between 65°C and 88°C (physiological range); however, very low pH appears to influence infection. Lastly, for the virus-host pairs tested the Fusellovirus-Sulfolobus system appears to exhibit host-advantage. This work provides a foundation for understanding Fusellovirus biology and virus-host coevolution in extreme ecosystems.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA