Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Oncol ; 17(7): 1192-1211, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37195379

RESUMO

Faithful and timely repair of DNA double-strand breaks (DSBs) is fundamental for the maintenance of genomic integrity. Here, we demonstrate that the meiotic recombination co-factor MND1 facilitates the repair of DSBs in somatic cells. We show that MND1 localizes to DSBs, where it stimulates DNA repair through homologous recombination (HR). Importantly, MND1 is not involved in the response to replication-associated DSBs, implying that it is dispensable for HR-mediated repair of one-ended DSBs. Instead, we find that MND1 specifically plays a role in the response to two-ended DSBs that are induced by irradiation (IR) or various chemotherapeutic drugs. Surprisingly, we find that MND1 is specifically active in G2 phase, whereas it only marginally affects repair during S phase. MND1 localization to DSBs is dependent on resection of the DNA ends and seemingly occurs through direct binding of MND1 to RAD51-coated ssDNA. Importantly, the lack of MND1-driven HR repair directly potentiates the toxicity of IR-induced damage, which could open new possibilities for therapeutic intervention, specifically in HR-proficient tumors.


Assuntos
Reparo do DNA , Recombinação Homóloga , Humanos , Reparo do DNA/genética , Recombinação Homóloga/genética , Quebras de DNA de Cadeia Dupla , Reparo de DNA por Recombinação , Fase S , Proteínas de Ciclo Celular/metabolismo
2.
Nat Commun ; 13(1): 754, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35136067

RESUMO

The genome consists of regions of transcriptionally active euchromatin and more silent heterochromatin. We reveal that the formation of heterochromatin domains requires cohesin turnover on DNA. Stabilization of cohesin on DNA through depletion of its release factor WAPL leads to a near-complete loss of heterochromatin domains. We observe the opposite phenotype in cells deficient for subunits of the Mediator-CDK module, with an almost binary partition of the genome into dense H3K9me3 domains, and regions devoid of H3K9me3 spanning the rest of the genome. We suggest that the Mediator-CDK module might contribute to gene expression by limiting the formation of dense heterochromatin domains. WAPL deficiency prevents the formation of heterochromatin domains, and allows for gene expression even in the absence of the Mediator-CDK subunit MED12. We propose that cohesin and Mediator affect heterochromatin in different ways to enable the correct distribution of epigenetic marks, and thus to ensure proper gene expression.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Heterocromatina/metabolismo , Complexo Mediador/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Transporte/genética , Linhagem Celular , Sequenciamento de Cromatina por Imunoprecipitação , Epigênese Genética , Técnicas de Inativação de Genes , Humanos , Complexo Mediador/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogênicas/genética , RNA-Seq , Coesinas
3.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33443154

RESUMO

The journey from plasma membrane to nuclear pore is a critical step in the lifecycle of DNA viruses, many of which must successfully deposit their genomes into the nucleus for replication. Viral capsids navigate this vast distance through the coordinated hijacking of a number of cellular host factors, many of which remain unknown. We performed a gene-trap screen in haploid cells to identify host factors for adenovirus (AdV), a DNA virus that can cause severe respiratory illness in immune-compromised individuals. This work identified Mindbomb 1 (MIB1), an E3 ubiquitin ligase involved in neurodevelopment, as critical for AdV infectivity. In the absence of MIB1, we observed that viral capsids successfully traffic to the proximity of the nucleus but ultimately fail to deposit their genomes within. The capacity of MIB1 to promote AdV infection was dependent on its ubiquitination activity, suggesting that MIB1 may mediate proteasomal degradation of one or more negative regulators of AdV infection. Employing complementary proteomic approaches to characterize proteins proximal to MIB1 upon AdV infection and differentially ubiquitinated in the presence or absence of MIB1, we observed an intersection between MIB1 and ribonucleoproteins (RNPs) largely unexplored in mammalian cells. This work uncovers yet another way that viruses utilize host cell machinery for their own replication, highlighting a potential target for therapeutic interventions that counter AdV infection.


Assuntos
Infecções por Adenoviridae/metabolismo , Adenoviridae/genética , Ubiquitina-Proteína Ligases/metabolismo , Células A549 , Infecções por Adenoviridae/genética , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Poro Nuclear/metabolismo , Ligação Proteica , Proteômica , Ribonucleoproteínas/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Ubiquitinação , Vírion/metabolismo , Replicação Viral/fisiologia
4.
Immunity ; 54(1): 132-150.e9, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33271119

RESUMO

HLA class I (HLA-I) glycoproteins drive immune responses by presenting antigens to cognate CD8+ T cells. This process is often hijacked by tumors and pathogens for immune evasion. Because options for restoring HLA-I antigen presentation are limited, we aimed to identify druggable HLA-I pathway targets. Using iterative genome-wide screens, we uncovered that the cell surface glycosphingolipid (GSL) repertoire determines effective HLA-I antigen presentation. We show that absence of the protease SPPL3 augmented B3GNT5 enzyme activity, resulting in upregulation of surface neolacto-series GSLs. These GSLs sterically impeded antibody and receptor interactions with HLA-I and diminished CD8+ T cell activation. Furthermore, a disturbed SPPL3-B3GNT5 pathway in glioma correlated with decreased patient survival. We show that the immunomodulatory effect could be reversed through GSL synthesis inhibition using clinically approved drugs. Overall, our study identifies a GSL signature that inhibits immune recognition and represents a potential therapeutic target in cancer, infection, and autoimmunity.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Linfócitos T CD8-Positivos/imunologia , Glioma/imunologia , Glicoesfingolipídeos/metabolismo , Glicosiltransferases/metabolismo , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoterapia/métodos , Apresentação de Antígeno , Ácido Aspártico Endopeptidases/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioma/mortalidade , Glicoesfingolipídeos/imunologia , Antígenos HLA/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Ativação Linfocitária , Transdução de Sinais , Análise de Sobrevida , Evasão Tumoral
5.
Sci Signal ; 13(649)2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32934076

RESUMO

Forward genetic screens in mammalian cell lines, such as RNAi and CRISPR-Cas9 screens, have made major contributions to the elucidation of diverse signaling pathways. Here, we exploited human haploid cells as a robust comparative screening platform and report a set of quantitative forward genetic screens for identifying regulatory mechanisms of mTORC1 signaling, a key growth control pathway that senses diverse metabolic states. Selected chemical and genetic perturbations in this screening platform, including rapamycin treatment and genetic ablation of the Ragulator subunit LAMTOR4, revealed the known core mTORC1 regulatory signaling complexes and the intimate interplay of the mTORC1 pathway with lysosomal function, validating the approach. In addition, we identified a differential requirement for LAMTOR4 and LAMTOR5 in regulating the mTORC1 pathway under fed and starved conditions. Furthermore, we uncovered a previously unknown "synthetic-sick" interaction between the tumor suppressor folliculin and LAMTOR4, which may have therapeutic implications in cancer treatment. Together, our study demonstrates the use of iterative "perturb and observe" genetic screens to uncover regulatory mechanisms driving complex mammalian signaling networks.


Assuntos
Retroalimentação Fisiológica , Testes Genéticos/métodos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HEK293 , Haploidia , Células HeLa , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Mutação , Proteínas Proto-Oncogênicas/genética , Proteínas Supressoras de Tumor/genética
6.
Cell Rep ; 32(8): 108068, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32846126

RESUMO

Using genome-wide radiogenetic profiling, we functionally dissect vulnerabilities of cancer cells to ionizing radiation (IR). We identify ERCC6L2 as a major determinant of IR response, together with classical DNA damage response genes and members of the recently identified shieldin and CTC1-STN1-TEN1 (CST) complexes. We show that ERCC6L2 contributes to non-homologous end joining (NHEJ), and it may exert this function through interactions with SFPQ. In addition to causing radiosensitivity, ERCC6L2 loss restores DNA end resection and partially rescues homologous recombination (HR) in BRCA1-deficient cells. As a consequence, ERCC6L2 deficiency confers resistance to poly (ADP-ribose) polymerase (PARP) inhibition in tumors deficient for both BRCA1 and p53. Moreover, we show that ERCC6L2 mutations are found in human tumors and correlate with a better overall survival in patients treated with radiotherapy (RT); this finding suggests that ERCC6L2 is a predictive biomarker of RT response.


Assuntos
Reparo do DNA por Junção de Extremidades/efeitos da radiação , DNA Helicases/metabolismo , Animais , Humanos , Camundongos
7.
J Virol ; 93(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30996093

RESUMO

Vaccinia virus is a promising viral vaccine and gene delivery candidate and has historically been used as a model to study poxvirus-host cell interactions. We employed a genome-wide insertional mutagenesis approach in human haploid cells to identify host factors crucial for vaccinia virus infection. A library of mutagenized HAP1 cells was exposed to modified vaccinia virus Ankara (MVA). Deep-sequencing analysis of virus-resistant cells identified host factors involved in heparan sulfate synthesis, Golgi organization, and vesicular protein trafficking. We validated EXT1, TM9SF2, and TMED10 (TMP21/p23/p24δ) as important host factors for vaccinia virus infection. The critical roles of EXT1 in heparan sulfate synthesis and vaccinia virus infection were confirmed. TM9SF2 was validated as a player mediating heparan sulfate expression, explaining its contribution to vaccinia virus infection. In addition, TMED10 was found to be crucial for virus-induced plasma membrane blebbing and phosphatidylserine-induced macropinocytosis, presumably by regulating the cell surface expression of the TAM receptor Axl.IMPORTANCE Poxviruses are large DNA viruses that can infect a wide range of host species. A number of these viruses are clinically important to humans, including variola virus (smallpox) and vaccinia virus. Since the eradication of smallpox, zoonotic infections with monkeypox virus and cowpox virus are emerging. Additionally, poxviruses can be engineered to specifically target cancer cells and are used as a vaccine vector against tuberculosis, influenza, and coronaviruses. Poxviruses rely on host factors for most stages of their life cycle, including attachment to the cell and entry. These host factors are crucial for virus infectivity and host cell tropism. We used a genome-wide knockout library of host cells to identify host factors necessary for vaccinia virus infection. We confirm a dominant role for heparin sulfate in mediating virus attachment. Additionally, we show that TMED10, previously not implicated in virus infections, facilitates virus uptake by modulating the cellular response to phosphatidylserine.


Assuntos
Haploidia , Heparitina Sulfato/genética , Heparitina Sulfato/isolamento & purificação , Pinocitose/fisiologia , Vaccinia virus/genética , Vaccinia virus/metabolismo , Vacínia/virologia , Proteínas de Transporte Vesicular/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Vírus da Varíola Bovina/genética , Vírus de DNA , Técnicas de Inativação de Genes , Testes Genéticos , Complexo de Golgi , Células HEK293 , Células HeLa , Heparitina Sulfato/metabolismo , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Humanos , Proteínas de Membrana , Monkeypox virus/genética , N-Acetilglucosaminiltransferases , Fosfatidilserinas/metabolismo , Poxviridae/genética , Ligação Viral
8.
Science ; 362(6419): 1171-1177, 2018 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-30442766

RESUMO

In genetic screens aimed at understanding drug resistance mechanisms in chronic myeloid leukemia cells, inactivation of the cullin 3 adapter protein-encoding leucine zipper-like transcription regulator 1 (LZTR1) gene led to enhanced mitogen-activated protein kinase (MAPK) pathway activity and reduced sensitivity to tyrosine kinase inhibitors. Knockdown of the Drosophila LZTR1 ortholog CG3711 resulted in a Ras-dependent gain-of-function phenotype. Endogenous human LZTR1 associates with the main RAS isoforms. Inactivation of LZTR1 led to decreased ubiquitination and enhanced plasma membrane localization of endogenous KRAS (V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog). We propose that LZTR1 acts as a conserved regulator of RAS ubiquitination and MAPK pathway activation. Because LZTR1 disease mutations failed to revert loss-of-function phenotypes, our findings provide a molecular rationale for LZTR1 involvement in a variety of inherited and acquired human disorders.


Assuntos
Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Fatores de Transcrição/fisiologia , Ubiquitinação , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Drosophila melanogaster , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas de Fusão bcr-abl/antagonistas & inibidores , Mutação com Ganho de Função , Técnicas de Silenciamento de Genes , Humanos , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/epidemiologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Mutação com Perda de Função , Sistema de Sinalização das MAP Quinases/genética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Piridazinas/farmacologia , Piridazinas/uso terapêutico , Transdução de Sinais , Fatores de Transcrição/genética , Ubiquitinação/genética
9.
Cell Host Microbe ; 23(5): 636-643.e5, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29681460

RESUMO

Human type A Enteroviruses (EV-As) cause diseases ranging from hand-foot-and-mouth disease to poliomyelitis-like disease. Although cellular receptors are identified for some EV-As, they remain elusive for the majority of EV-As. We identify the cell surface molecule KREMEN1 as an entry receptor for coxsackievirus A10 (CV-A10). Whereas loss of KREMEN1 renders cells resistant to CV-A10 infection, KREMEN1 overexpression enhances CV-A10 binding to the cell surface and increases susceptibility to infection, indicating that KREMEN1 is a rate-limiting factor for CV-A10 infection. Furthermore, the extracellular domain of KREMEN1 binds CV-A10 and functions as a neutralizing agent during infection. Kremen-deficient mice are resistant to CV-A10-induced lethal paralysis, emphasizing the relevance of Kremen for infection in vivo. KREMEN1 is also essential for infection by a phylogenetic and pathogenic related group of EV-As. Collectively these findings highlight the importance of KREMEN1 for these emerging pathogens and its potential as an antiviral therapeutic target.


Assuntos
Enterovirus Humano A/metabolismo , Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/metabolismo , Proteínas de Membrana/metabolismo , Internalização do Vírus , Animais , Antígenos de Superfície , Linhagem Celular , Linhagem Celular Tumoral , Enterovirus/patogenicidade , Infecções por Enterovirus/imunologia , Infecções por Enterovirus/virologia , Feminino , Técnicas de Inativação de Genes , Células HCT116 , Células HEK293 , Doença de Mão, Pé e Boca/virologia , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos , Mutagênese , Filogenia , Domínios Proteicos
10.
Mol Oncol ; 12(6): 953-971, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29689640

RESUMO

The absence of biomarkers to accurately predict anticancer therapy response remains a major obstacle in clinical oncology. We applied a genome-wide loss-of-function screening approach in human haploid cells to characterize genetic vulnerabilities to classical microtubule-targeting agents. Using docetaxel and vinorelbine, two well-established chemotherapeutic agents, we sought to identify genetic alterations sensitizing human HAP1 cells to these drugs. Despite the fact that both drugs act on microtubules, a set of distinct genes were identified whose disruption affects drug sensitivity. For docetaxel, this included a number of genes with a function in mitosis, while for vinorelbine we identified inactivation of FBXW7, RB1, and NF2, three frequently mutated tumor suppressor genes, as sensitizing factors. We validated these genes using independent knockout clones and confirmed FBXW7 as an important regulator of the mitotic spindle assembly. Upon FBXW7 depletion, vinorelbine treatment led to decreased survival of cells due to defective mitotic progression and subsequent mitotic catastrophe. We show that haploid insertional mutagenesis screens are a useful tool to study genetic vulnerabilities to classical chemotherapeutic drugs by identifying thus far unknown sensitivity factors. These results provide a rationale for investigating patient response to vinca alkaloid-based anticancer treatment in relation to the mutational status of these three tumor suppressor genes, and could in the future lead to the establishment of novel predictive biomarkers or suggest new drug combinations based on molecular mechanisms of drug sensitivity.


Assuntos
Testes Genéticos , Haploidia , Microtúbulos/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proteína 7 com Repetições F-Box-WD/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Supressores de Tumor , Genoma Humano , Humanos , Microtúbulos/efeitos dos fármacos , Mitose/efeitos dos fármacos , Morfolinas/farmacologia , Mutagênese Insercional/genética , Mutação/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Purinas/farmacologia , Vinorelbina/farmacologia
11.
Cell Host Microbe ; 22(5): 688-696.e5, 2017 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-29120745

RESUMO

Arenaviruses cause fatal hemorrhagic disease in humans. Old World arenavirus glycoproteins (GPs) mainly engage α-dystroglycan as a cell-surface receptor, while New World arenaviruses hijack transferrin receptor. However, the Lujo virus (LUJV) GP does not cluster with New or Old World arenaviruses. Using a recombinant vesicular stomatitis virus containing LUJV GP as its sole attachment and fusion protein (VSV-LUJV), we demonstrate that infection is independent of known arenavirus receptor genes. A genome-wide haploid genetic screen identified the transmembrane protein neuropilin 2 (NRP2) and tetraspanin CD63 as factors for LUJV GP-mediated infection. LUJV GP binds the N-terminal domain of NRP2, while CD63 stimulates pH-activated LUJV GP-mediated membrane fusion. Overexpression of NRP2 or its N-terminal domain enhances VSV-LUJV infection, and cells lacking NRP2 are deficient in wild-type LUJV infection. These findings uncover this distinct set of host cell entry factors in LUJV infection and are attractive focus points for therapeutic intervention.


Assuntos
Lujo virus/fisiologia , Neuropilina-2/metabolismo , Tetraspanina 30/metabolismo , Proteínas Virais de Fusão/metabolismo , Proteínas Virais/metabolismo , Internalização do Vírus , Proteínas de Transporte , Linhagem Celular , Interações Hospedeiro-Patógeno/fisiologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Lujo virus/genética , Lujo virus/patogenicidade , Domínios e Motivos de Interação entre Proteínas , Receptores de Superfície Celular/metabolismo , Receptores da Transferrina , Proteínas Virais de Fusão/genética , Proteínas Virais/genética
12.
Cell Host Microbe ; 22(4): 460-470.e5, 2017 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-29024641

RESUMO

Respiratory and arthropod-borne viral infections are a global threat due to the lack of effective antivirals and vaccines. A potential strategy is to target host proteins required for viruses but non-essential for the host. To identify such proteins, we performed a genome-wide knockout screen in human haploid cells and identified the calcium pump SPCA1. SPCA1 is required by viruses from the Paramyxoviridae, Flaviviridae, and Togaviridae families, including measles, dengue, West Nile, Zika, and chikungunya viruses. Calcium transport activity is required for SPCA1 to promote virus spread. SPCA1 regulates proteases within the trans-Golgi network that require calcium for their activity and are critical for virus glycoprotein maturation. Consistent with these findings, viral glycoproteins fail to mature in SPCA1-deficient cells preventing viral spread, which is evident even in cells with partial loss of SPCA1. Thus, SPCA1 is an attractive antiviral host target for a broad spectrum of established and emerging viral infections.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Cálcio/metabolismo , Flaviviridae/fisiologia , Interações Hospedeiro-Patógeno , Paramyxoviridae/fisiologia , Togaviridae/fisiologia , Proteínas Virais/metabolismo , Células A549 , Animais , ATPases Transportadoras de Cálcio/genética , Chlorocebus aethiops , Feminino , Técnicas de Inativação de Genes , Estudo de Associação Genômica Ampla , Haploidia , Células HeLa , Humanos , Masculino , Células Vero , Proteínas Virais/genética , Rede trans-Golgi/enzimologia
13.
Nature ; 546(7657): 307-311, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-28562590

RESUMO

As key executers of biological functions, the activity and abundance of proteins are subjected to extensive regulation. Deciphering the genetic architecture underlying this regulation is critical for understanding cellular signalling events and responses to environmental cues. Using random mutagenesis in haploid human cells, we apply a sensitive approach to directly couple genomic mutations to protein measurements in individual cells. Here we use this to examine a suite of cellular processes, such as transcriptional induction, regulation of protein abundance and splicing, signalling cascades (mitogen-activated protein kinase (MAPK), G-protein-coupled receptor (GPCR), protein kinase B (AKT), interferon, and Wingless and Int-related protein (WNT) pathways) and epigenetic modifications (histone crotonylation and methylation). This scalable, sequencing-based procedure elucidates the genetic landscapes that control protein states, identifying genes that cause very narrow phenotypic effects and genes that lead to broad phenotypic consequences. The resulting genetic wiring map identifies the E3-ligase substrate adaptor KCTD5 (ref. 1) as a negative regulator of the AKT pathway, a key signalling cascade frequently deregulated in cancer. KCTD5-deficient cells show elevated levels of phospho-AKT at S473 that could not be attributed to effects on canonical pathway components. To reveal the genetic requirements for this phenotype, we iteratively analysed the regulatory network linked to AKT activity in the knockout background. This genetic modifier screen exposes suppressors of the KCTD5 phenotype and mechanistically demonstrates that KCTD5 acts as an off-switch for GPCR signalling by triggering proteolysis of Gßγ heterodimers dissociated from the Gα subunit. Although biological networks have previously been constructed on the basis of gene expression, protein-protein associations, or genetic interaction profiles, we foresee that the approach described here will enable the generation of a comprehensive genetic wiring map for human cells on the basis of quantitative protein states.


Assuntos
Canais de Potássio/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/genética , Análise de Célula Única/métodos , Células Cultivadas , Haploidia , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Interferons/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutagênese , Fenótipo , Fosforilação/genética , Canais de Potássio/deficiência , Canais de Potássio/genética , Proteólise , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Via de Sinalização Wnt
14.
Cell ; 169(4): 693-707.e14, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28475897

RESUMO

The spatial organization of chromosomes influences many nuclear processes including gene expression. The cohesin complex shapes the 3D genome by looping together CTCF sites along chromosomes. We show here that chromatin loop size can be increased and that the duration with which cohesin embraces DNA determines the degree to which loops are enlarged. Cohesin's DNA release factor WAPL restricts this loop extension and also prevents looping between incorrectly oriented CTCF sites. We reveal that the SCC2/SCC4 complex promotes the extension of chromatin loops and the formation of topologically associated domains (TADs). Our data support the model that cohesin structures chromosomes through the processive enlargement of loops and that TADs reflect polyclonal collections of loops in the making. Finally, we find that whereas cohesin promotes chromosomal looping, it rather limits nuclear compartmentalization. We conclude that the balanced activity of SCC2/SCC4 and WAPL enables cohesin to correctly structure chromosomes.


Assuntos
Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Cromatina/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Acetiltransferases/metabolismo , Fator de Ligação a CCCTC , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA , Elongases de Ácidos Graxos , Edição de Genes , Humanos , Complexos Multiproteicos/metabolismo , Proteínas Repressoras/metabolismo , Coesinas
15.
Nature ; 541(7637): 412-416, 2017 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-28077878

RESUMO

Picornaviruses are a leading cause of human and veterinary infections that result in various diseases, including polio and the common cold. As archetypical non-enveloped viruses, their biology has been extensively studied. Although a range of different cell-surface receptors are bound by different picornaviruses, it is unclear whether common host factors are needed for them to reach the cytoplasm. Using genome-wide haploid genetic screens, here we identify the lipid-modifying enzyme PLA2G16 (refs 8, 9, 10, 11) as a picornavirus host factor that is required for a previously unknown event in the viral life cycle. We find that PLA2G16 functions early during infection, enabling virion-mediated genome delivery into the cytoplasm, but not in any virion-assigned step, such as cell binding, endosomal trafficking or pore formation. To resolve this paradox, we screened for suppressors of the ΔPLA2G16 phenotype and identified a mechanism previously implicated in the clearance of intracellular bacteria. The sensor of this mechanism, galectin-8 (encoded by LGALS8), detects permeated endosomes and marks them for autophagic degradation, whereas PLA2G16 facilitates viral genome translocation and prevents clearance. This study uncovers two competing processes triggered by virus entry: activation of a pore-activated clearance pathway and recruitment of a phospholipase to enable genome release.


Assuntos
Citoplasma/virologia , Genoma Viral , Fatores Celulares Derivados do Hospedeiro/metabolismo , Fosfolipases A2 Independentes de Cálcio/metabolismo , Picornaviridae/genética , Picornaviridae/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Internalização do Vírus , Animais , Autofagia , Transporte Biológico , Linhagem Celular , Citoplasma/genética , Endossomos/metabolismo , Feminino , Galectinas/genética , Galectinas/metabolismo , Fatores Celulares Derivados do Hospedeiro/deficiência , Fatores Celulares Derivados do Hospedeiro/genética , Humanos , Masculino , Camundongos , Mutação , Fenótipo , Fosfolipases A2 Independentes de Cálcio/deficiência , Fosfolipases A2 Independentes de Cálcio/genética , Supressão Genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Vírion/genética , Vírion/metabolismo , Replicação Viral
16.
Proc Natl Acad Sci U S A ; 112(49): 15160-5, 2015 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-26598692

RESUMO

The Wee1 cell cycle checkpoint kinase prevents premature mitotic entry by inhibiting cyclin-dependent kinases. Chemical inhibitors of Wee1 are currently being tested clinically as targeted anticancer drugs. Wee1 inhibition is thought to be preferentially cytotoxic in p53-defective cancer cells. However, TP53 mutant cancers do not respond consistently to Wee1 inhibitor treatment, indicating the existence of genetic determinants of Wee1 inhibitor sensitivity other than TP53 status. To optimally facilitate patient selection for Wee1 inhibition and uncover potential resistance mechanisms, identification of these currently unknown genes is necessary. The aim of this study was therefore to identify gene mutations that determine Wee1 inhibitor sensitivity. We performed a genome-wide unbiased functional genetic screen in TP53 mutant near-haploid KBM-7 cells using gene-trap insertional mutagenesis. Insertion site mapping of cells that survived long-term Wee1 inhibition revealed enrichment of G1/S regulatory genes, including SKP2, CUL1, and CDK2. Stable depletion of SKP2, CUL1, or CDK2 or chemical Cdk2 inhibition rescued the γ-H2AX induction and abrogation of G2 phase as induced by Wee1 inhibition in breast and ovarian cancer cell lines. Remarkably, live cell imaging showed that depletion of SKP2, CUL1, or CDK2 did not rescue the Wee1 inhibition-induced karyokinesis and cytokinesis defects. These data indicate that the activity of the DNA replication machinery, beyond TP53 mutation status, determines Wee1 inhibitor sensitivity, and could serve as a selection criterion for Wee1-inhibitor eligible patients. Conversely, loss of the identified S-phase genes could serve as a mechanism of acquired resistance, which goes along with development of severe genomic instability.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Fase G1 , Haploidia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Fase S , Fase G1/genética , Humanos , Fase S/genética
17.
EMBO J ; 34(24): 2993-3008, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26530471

RESUMO

Although platinum-based drugs are widely used chemotherapeutics for cancer treatment, the determinants of tumor cell responsiveness remain poorly understood. We show that the loss of subunits LRRC8A and LRRC8D of the heteromeric LRRC8 volume-regulated anion channels (VRACs) increased resistance to clinically relevant cisplatin/carboplatin concentrations. Under isotonic conditions, about 50% of cisplatin uptake depended on LRRC8A and LRRC8D, but neither on LRRC8C nor on LRRC8E. Cell swelling strongly enhanced LRRC8-dependent cisplatin uptake, bolstering the notion that cisplatin enters cells through VRAC. LRRC8A disruption also suppressed drug-induced apoptosis independently from drug uptake, possibly by impairing VRAC-dependent apoptotic cell volume decrease. Hence, by mediating cisplatin uptake and facilitating apoptosis, VRAC plays a dual role in the cellular drug response. Incorporation of the LRRC8D subunit into VRAC substantially increased its permeability for cisplatin and the cellular osmolyte taurine, indicating that LRRC8 proteins form the channel pore. Our work suggests that LRRC8D-containing VRACs are crucial for cell volume regulation by an important organic osmolyte and may influence cisplatin/carboplatin responsiveness of tumors.


Assuntos
Antineoplásicos/farmacologia , Carboplatina/farmacologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Proteínas de Membrana/metabolismo , Apoptose , Tamanho Celular , Células HCT116 , Células HEK293 , Humanos , Proteínas de Membrana/genética , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo
18.
Nat Chem Biol ; 10(9): 768-773, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25064833

RESUMO

Genotoxic chemotherapy is the most common cancer treatment strategy. However, its untargeted generic DNA-damaging nature and associated systemic cytotoxicity greatly limit its therapeutic applications. Here, we used a haploid genetic screen in human cells to discover an absolute dependency of the clinically evaluated anticancer compound YM155 on solute carrier family member 35 F2 (SLC35F2), an uncharacterized member of the solute carrier protein family that is highly expressed in a variety of human cancers. YM155 generated DNA damage through intercalation, which was contingent on the expression of SLC35F2 and its drug-importing activity. SLC35F2 expression and YM155 sensitivity correlated across a panel of cancer cell lines, and targeted genome editing verified SLC35F2 as the main determinant of YM155-mediated DNA damage toxicity in vitro and in vivo. These findings suggest a new route to targeted DNA damage by exploiting tumor and patient-specific import of YM155.


Assuntos
Dano ao DNA/efeitos dos fármacos , Imidazóis/farmacologia , Substâncias Intercalantes/farmacologia , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Naftoquinonas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular , Clonagem Molecular , Ensaio Cometa , Genoma Humano/efeitos dos fármacos , Genoma Humano/genética , Haploidia , Humanos , Imidazóis/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos SCID , Naftoquinonas/metabolismo , RNA Neoplásico/química , RNA Neoplásico/genética
19.
Nat Methods ; 10(10): 965-71, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24161985

RESUMO

Knockout collections are invaluable tools for studying model organisms such as yeast. However, there are no large-scale knockout collections of human cells. Using gene-trap mutagenesis in near-haploid human cells, we established a platform to generate and isolate individual 'gene-trapped cells' and used it to prepare a collection of human cell lines carrying single gene-trap insertions. In most cases, the insertion can be reversed. This growing library covers 3,396 genes, one-third of the expressed genome, is DNA-barcoded and allows systematic screens for a wide variety of cellular phenotypes. We examined cellular responses to TNF-α, TGF-ß, IFN-γ and TNF-related apoptosis-inducing ligand (TRAIL), to illustrate the value of this unique collection of isogenic human cell lines.


Assuntos
Biblioteca Gênica , Haploidia , Mutagênese Insercional/métodos , Genética Reversa/métodos , Linhagem Celular Tumoral , Genoma Humano , Humanos , Dados de Sequência Molecular
20.
Science ; 340(6131): 479-83, 2013 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-23519211

RESUMO

Glycosylated α-dystroglycan (α-DG) serves as cellular entry receptor for multiple pathogens, and defects in its glycosylation cause hereditary Walker-Warburg syndrome (WWS). At least eight proteins are critical to glycosylate α-DG, but many genes mutated in WWS remain unknown. To identify modifiers of α-DG, we performed a haploid screen for Lassa virus entry, a hemorrhagic fever virus causing thousands of deaths annually that hijacks glycosylated α-DG to enter cells. In complementary screens, we profiled cells for absence of α-DG carbohydrate chains or biochemically related glycans. This revealed virus host factors and a suite of glycosylation units, including all known Walker-Warburg genes and five additional factors critical for the modification of α-DG. Our findings accentuate the complexity of this posttranslational feature and point out genes defective in dystroglycanopathies.


Assuntos
Distroglicanas/metabolismo , Interações Hospedeiro-Patógeno/genética , Febre Lassa/genética , Vírus Lassa/fisiologia , Proteínas de Membrana/genética , Proteoma/metabolismo , Internalização do Vírus , Síndrome de Walker-Warburg/genética , Sequência de Aminoácidos , Linhagem Celular , Feminino , Glicosilação , Haploidia , Humanos , Lactente , Febre Lassa/virologia , Masculino , Dados de Sequência Molecular , Mutação , Linhagem , Pentosiltransferases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA