Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Biol ; 22(1): e3002406, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38227562

RESUMO

Breast tumours are embedded in a collagen I-rich extracellular matrix (ECM) network, where nutrients are scarce due to limited blood flow and elevated tumour growth. Metabolic adaptation is required for cancer cells to endure these conditions. Here, we demonstrated that the presence of ECM supported the growth of invasive breast cancer cells, but not non-transformed mammary epithelial cells, under amino acid starvation, through a mechanism that required macropinocytosis-dependent ECM uptake. Importantly, we showed that this behaviour was acquired during carcinoma progression. ECM internalisation, followed by lysosomal degradation, contributed to the up-regulation of the intracellular levels of several amino acids, most notably tyrosine and phenylalanine. This resulted in elevated tyrosine catabolism on ECM under starvation, leading to increased fumarate levels, potentially feeding into the tricarboxylic acid (TCA) cycle. Interestingly, this pathway was required for ECM-dependent cell growth and invasive cell migration under amino acid starvation, as the knockdown of p-hydroxyphenylpyruvate hydroxylase-like protein (HPDL), the third enzyme of the pathway, opposed cell growth and motility on ECM in both 2D and 3D systems, without affecting cell proliferation on plastic. Finally, high HPDL expression correlated with poor prognosis in breast cancer patients. Collectively, our results highlight that the ECM in the tumour microenvironment (TME) represents an alternative source of nutrients to support cancer cell growth by regulating phenylalanine and tyrosine metabolism.


Assuntos
Aminoácidos , Neoplasias da Mama , Humanos , Feminino , Aminoácidos/metabolismo , Neoplasias da Mama/metabolismo , Matriz Extracelular/metabolismo , Tirosina/metabolismo , Fenilalanina , Microambiente Tumoral
2.
Matrix Biol ; 118: 110-128, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36924903

RESUMO

Imbalance of collagen I expression results in severe pathologies. Apart from activation by the TGFß-receptor/Smad pathway, control of collagen I expression remains poorly understood. Here, we used human dermal fibroblasts expressing a mCherry fluorescent protein driven by endogenous COL1A1 promoter to functionally screen the kinome and phosphatome. We identify 8 negative regulators, revealing that collagen is under tonic repression. The cell surface receptor BDKRB2 represses collagen I and other pro-fibrotic genes. Interestingly, it also promotes other basal membrane ECM genes. This function is independent of the natural ligand, bradykinin, and of SMAD2/3 factors, instead requiring constant ERK1/2 repression. TGFß stimulation induces rapid BDKRB2 transcriptional downregulation. Human fibrotic fibroblasts have reduced BDKRB2 levels and enhancing its expression in keloid fibroblasts represses COL1A1. We propose that tonic signalling by BDKRB2 prevents collagen overproduction in skin fibroblasts.


Assuntos
Colágeno Tipo I , Pele , Humanos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Pele/metabolismo , Colágeno/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Receptores da Bradicinina/metabolismo
3.
Elife ; 102021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34870592

RESUMO

The Src tyrosine kinase controls cancer-critical protein glycosylation through Golgi to ER relocation of GALNTs enzymes. How Src induces this trafficking event is unknown. Golgi to ER transport depends on the GTP exchange factor (GEF) GBF1 and small GTPase Arf1. Here, we show that Src induces the formation of tubular transport carriers containing GALNTs. The kinase phosphorylates GBF1 on 10 tyrosine residues; two of them, Y876 and Y898, are located near the C-terminus of the Sec7 GEF domain. Their phosphorylation promotes GBF1 binding to the GTPase; molecular modeling suggests partial melting of the Sec7 domain and intramolecular rearrangement. GBF1 mutants defective for these rearrangements prevent binding, carrier formation, and GALNTs relocation, while phosphomimetic GBF1 mutants induce tubules. In sum, Src promotes GALNTs relocation by promoting GBF1 binding to Arf1. Based on residue conservation, similar regulation of GEF-Arf complexes by tyrosine phosphorylation could be a conserved and widespread mechanism.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/genética , Quinases da Família src/genética , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Fosforilação , Transporte Proteico , Quinases da Família src/metabolismo
5.
Sci Rep ; 10(1): 19723, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-33184327

RESUMO

Type I collagen is a key protein of most connective tissue and its up-regulation is required for wound healing but is also involved in fibrosis. Control of expression of this collagen remains poorly understood apart from Transforming Growth Factor beta (TGF-ß1)-mediated induction. To generate a sensitive, practical, robust, image-based high-throughput-compatible reporter system, we genetically inserted a short-lived fluorescence reporter downstream of the endogenous type I collagen (COL1A1) promoter in skin fibroblasts. Using a variety of controls, we demonstrate that the cell line faithfully reports changes in type I collagen expression with at least threefold enhanced sensitivity compared to endogenous collagen monitoring. We use this assay to test the potency of anti-fibrotic compounds and screen siRNAs for regulators of TGF-ß1-induced type I collagen expression. We propose our reporter cell line, Red-COLA1, as a new efficient tool to study type I collagen transcriptional regulation.


Assuntos
Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Fibrose/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Indóis/farmacologia , Proteínas Luminescentes/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Células Cultivadas , Colágeno/metabolismo , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Fibroblastos/citologia , Fibrose/tratamento farmacológico , Fibrose/patologia , Ensaios de Triagem em Larga Escala , Humanos , Proteínas Luminescentes/genética , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Transcrição Gênica , Fator de Crescimento Transformador beta1/genética , Proteína Vermelha Fluorescente
6.
Nat Cell Biol ; 22(11): 1371-1381, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33077910

RESUMO

Tumour growth and invasiveness require extracellular matrix (ECM) degradation and are stimulated by the GALA pathway, which induces protein O-glycosylation in the endoplasmic reticulum (ER). ECM degradation requires metalloproteases, but whether other enzymes are required is unclear. Here, we show that GALA induces the glycosylation of the ER-resident calnexin (Cnx) in breast and liver cancer. Glycosylated Cnx and its partner ERp57 are trafficked to invadosomes, which are sites of ECM degradation. We find that disulfide bridges are abundant in connective and liver ECM. Cell surface Cnx-ERp57 complexes reduce these extracellular disulfide bonds and are essential for ECM degradation. In vivo, liver cancer cells but not hepatocytes display cell surface Cnx. Liver tumour growth and lung metastasis of breast and liver cancer cells are inhibited by anti-Cnx antibodies. These findings uncover a moonlighting function of Cnx-ERp57 at the cell surface that is essential for ECM breakdown and tumour development.


Assuntos
Neoplasias da Mama/enzimologia , Calnexina/metabolismo , Movimento Celular , Retículo Endoplasmático/enzimologia , Matriz Extracelular/metabolismo , Neoplasias Hepáticas/enzimologia , Neoplasias Pulmonares/enzimologia , Podossomos/enzimologia , Isomerases de Dissulfetos de Proteínas/metabolismo , Animais , Antineoplásicos Imunológicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Calnexina/antagonistas & inibidores , Linhagem Celular Tumoral , Retículo Endoplasmático/patologia , Matriz Extracelular/patologia , Feminino , Glicosilação , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Células NIH 3T3 , Invasividade Neoplásica , Podossomos/patologia , Transporte Proteico , Proteólise , Ensaios Antitumorais Modelo de Xenoenxerto , alfa-Galactosidase/metabolismo
7.
Sci Rep ; 10(1): 17762, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33082422

RESUMO

Biologics such as peptides and antibodies are a well-established class of therapeutics. However, their intracellular delivery remains problematic. In particular, methods to efficiently inhibit intra-nuclear targets are lacking. We previously described that Pseudomonas Exotoxin A reaches the nucleoplasm via the endosomes-to-nucleus trafficking pathway. Here, we show that a non-toxic truncated form of PE can be coupled to peptides and efficiently reach the nucleoplasm. It can be used as a Peptide Nuclear Delivery Device (PNDD) to deliver polypeptidic cargos as large as Glutathione- S-transferase (GST) to the nucleus. PNDD1 is a fusion of PNDD to the c-myc inhibitor peptide H1. PNDD1 is able to inhibit c-Myc dependent transcription at nanomolar concentration. In contrast, H1 fused to various cell-penetrating peptides are active only in the micromolar range. PNDD1 attenuates cell proliferation and induces cell death in various tumor cell lines. In particular, several patient-derived Diffuse Large B-Cell Lymphomas cell lines die after exposure to PNDD1, while normal B-cells survive. Altogether, our data indicate that PNDD is a powerful tool to bring active cargo to the nucleus and PNDD1 could be the basis of a new therapy against lymphoma.


Assuntos
Núcleo Celular/metabolismo , Peptídeos Penetradores de Células/administração & dosagem , Transporte Proteico/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos
8.
Cells ; 8(8)2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31426451

RESUMO

Various growth factors and full-length cell surface receptors such as EGFR are translocated from the cell surface to the nucleoplasm, baffling cell biologists to the mechanisms and functions of this process. Elevated levels of nuclear EGFR correlate with poor prognosis in various cancers. In recent years, nuclear EGFR has been implicated in regulating gene transcription, cell proliferation and DNA damage repair. Different models have been proposed to explain how the receptors are transported into the nucleus. However, a clear consensus has yet to be reached. Recently, we described the nuclear envelope associated endosomes (NAE) pathway, which delivers EGFR from the cell surface to the nucleus. This pathway involves transport, docking and fusion of NAEs with the outer membrane of the nuclear envelope. EGFR is then presumed to be transported through the nuclear pore complex, extracted from membranes and solubilised. The SUN1/2 nuclear envelope proteins, Importin-beta, nuclear pore complex proteins and the Sec61 translocon have been implicated in the process. While this framework can explain the cell surface to nucleus traffic of EGFR and other cell surface receptors, it raises several questions that we consider in this review, together with implications for health and disease.


Assuntos
Endossomos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Nucleares/metabolismo , Canais de Translocação SEC/metabolismo , beta Carioferinas/metabolismo , Linhagem Celular , Endocitose , Receptores ErbB/metabolismo , Humanos
9.
PLoS One ; 14(3): e0214118, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30889231

RESUMO

The enzymes GALNTs add GalNAc sugar to Ser and Thr residues, forming the Tn glycan. GALNTs are activated by trafficking from Golgi to ER, a process driven by the Src kinase and negatively regulated by ERK8. This GALNTs activation (aka GALA) pathway induces high Tn levels and is a key driver of liver tumor growth. Recently, Tabak and colleagues have contested our previous data that EGF stimulation can induce GALNTs relocation. Here, we show that relocation induced by EGF is actually detectable in the very images acquired by Tabak et al. Furthermore, we show that over-expression of EGFR strongly enhances EGF-induced relocation and that EGFR appears required to drive relocation induced by ERK8 depletion. Direct co-localisation of GALNT with the ER marker Calnexin is observed after EGF stimulation. We furthermore propose that quantification of O-glycosylation of the ER resident protein PDIA4 provides a mean to quantify GALA independently of imaging. In sum, we demonstrate that the claimed non-reproducibility was due to experimental imaging conditions, that EGFR is indeed a driver of GALA and propose additional markers to facilitate the study of this pathway.


Assuntos
Retículo Endoplasmático/enzimologia , N-Acetilgalactosaminiltransferases/metabolismo , Retículo Endoplasmático/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glicosilação , Células HEK293 , Células HeLa , Humanos , N-Acetilgalactosaminiltransferases/genética , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo
10.
Glycobiology ; 28(8): 580-591, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29757379

RESUMO

Quiescin sulfhydryl oxidase 1 (QSOX1) catalyzes the formation of disulfide bonds in protein substrates. Unlike other enzymes with related activities, which are commonly found in the endoplasmic reticulum, QSOX1 is localized to the Golgi apparatus or secreted. QSOX1 is upregulated in quiescent fibroblast cells and secreted into the extracellular environment, where it contributes to extracellular matrix assembly. QSOX1 is also upregulated in adenocarcinomas, though the extent to which it is secreted in this context is currently unknown. To achieve a better understanding of factors that dictate QSOX1 localization and function, we aimed to determine how post-translational modifications affect QSOX1 trafficking and activity. We found a highly conserved N-linked glycosylation site to be required for QSOX1 secretion from fibroblasts and other cell types. Notably, QSOX1 lacking a glycan at this site arrives at the Golgi, suggesting that it passes endoplasmic reticulum quality control but is not further transported to the cell surface for secretion. The QSOX1 transmembrane segment is dispensable for Golgi localization and secretion, as fully luminal and transmembrane variants displayed the same trafficking behavior. This study provides a key example of the effect of glycosylation on Golgi exit and contributes to an understanding of late secretory sorting and quality control.


Assuntos
Fibroblastos/metabolismo , Complexo de Golgi/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismo , Linhagem Celular , Fibroblastos/citologia , Glicosilação , Complexo de Golgi/genética , Humanos , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/genética , Transporte Proteico/fisiologia
11.
Nat Commun ; 9(1): 2031, 2018 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-29795195

RESUMO

Invadosomes are F-actin-based structures involved in extracellular matrix degradation, cell invasion, and metastasis formation. Analyzing their proteome is crucial to decipher their molecular composition, to understand their mechanisms, and to find specific elements to target them. However, the specific analysis of invadosomes is challenging, because it is difficult to maintain their integrity during isolation. In addition, classical purification methods often suffer from contaminations, which may impair data validation. To ensure the specific identification of invadosome components, we here develop a method that combines laser microdissection and mass spectrometry, enabling the analysis of subcellular structures in their native state based on low amounts of input material. Using this combinatorial method, we show that invadosomes contain specific components of the translational machinery, in addition to known marker proteins. Moreover, functional validation reveals that protein translation activity is an inherent property of invadosomes, which is required to maintain invadosome structure and activity.


Assuntos
Podossomos/metabolismo , Biossíntese de Proteínas , Proteômica/métodos , RNA Mensageiro/metabolismo , Actinas/metabolismo , Animais , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão/métodos , Matriz Extracelular/metabolismo , Humanos , Microdissecção e Captura a Laser/métodos , Camundongos , Células NIH 3T3 , Neoplasias/diagnóstico , Neoplasias/patologia , Podossomos/patologia , Espectrometria de Massas em Tandem/métodos
12.
Breast Cancer Res ; 20(1): 22, 2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29566768

RESUMO

BACKGROUND: Phosphatase and tensin homolog (PTEN) is one of the most frequently inactivated tumor suppressors in breast cancer. While PTEN itself is not considered a druggable target, PTEN synthetic-sick or synthetic-lethal (PTEN-SSL) genes are potential drug targets in PTEN-deficient breast cancers. Therefore, with the aim of identifying potential targets for precision breast cancer therapy, we sought to discover PTEN-SSL genes present in a broad spectrum of breast cancers. METHODS: To discover broad-spectrum PTEN-SSL genes in breast cancer, we used a multi-step approach that started with (1) a genome-wide short interfering RNA (siRNA) screen of ~ 21,000 genes in a pair of isogenic human mammary epithelial cell lines, followed by (2) a short hairpin RNA (shRNA) screen of ~ 1200 genes focused on hits from the first screen in a panel of 11 breast cancer cell lines; we then determined reproducibility of hits by (3) identification of overlaps between our results and reanalyzed data from 3 independent gene-essentiality screens, and finally, for selected candidate PTEN-SSL genes we (4) confirmed PTEN-SSL activity using either drug sensitivity experiments in a panel of 19 cell lines or mutual exclusivity analysis of publicly available pan-cancer somatic mutation data. RESULTS: The screens (steps 1 and 2) and the reproducibility analysis (step 3) identified six candidate broad-spectrum PTEN-SSL genes (PIK3CB, ADAMTS20, AP1M2, HMMR, STK11, and NUAK1). PIK3CB was previously identified as PTEN-SSL, while the other five genes represent novel PTEN-SSL candidates. Confirmation studies (step 4) provided additional evidence that NUAK1 and STK11 have PTEN-SSL patterns of activity. Consistent with PTEN-SSL status, inhibition of the NUAK1 protein kinase by the small molecule drug HTH-01-015 selectively impaired viability in multiple PTEN-deficient breast cancer cell lines, while mutations affecting STK11 and PTEN were largely mutually exclusive across large pan-cancer data sets. CONCLUSIONS: Six genes showed PTEN-SSL patterns of activity in a large proportion of PTEN-deficient breast cancer cell lines and are potential specific vulnerabilities in PTEN-deficient breast cancer. Furthermore, the NUAK1 PTEN-SSL vulnerability identified by RNA interference techniques can be recapitulated and exploited using the small molecule kinase inhibitor HTH-01-015. Thus, NUAK1 inhibition may be an effective strategy for precision treatment of PTEN-deficient breast tumors.


Assuntos
Neoplasias da Mama/genética , PTEN Fosfo-Hidrolase/genética , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Repressoras/genética , Quinases Proteína-Quinases Ativadas por AMP , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Genoma Humano/genética , Genômica/métodos , Humanos , Glândulas Mamárias Humanas/metabolismo , Proteínas de Neoplasias/genética , PTEN Fosfo-Hidrolase/deficiência , RNA Interferente Pequeno/genética , Mutações Sintéticas Letais/genética
13.
Nat Commun ; 9(1): 100, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29311615

RESUMO

The repression of telomerase activity during cellular differentiation promotes replicative aging and functions as a physiological barrier for tumorigenesis in long-lived mammals, including humans. However, the underlying mechanisms remain largely unclear. Here we describe how miR-615-3p represses hTERT expression. mir-615-3p is located in an intron of the HOXC5 gene, a member of the highly conserved homeobox family of transcription factors controlling embryogenesis and development. Unexpectedly, we found that HoxC5 also represses hTERT expression by disrupting the long-range interaction between hTERT promoter and its distal enhancer. The 3'UTR of hTERT and its upstream enhancer region are well conserved in long-lived primates. Both mir-615-3p and HOXC5 are activated upon differentiation, which constitute a feed-forward loop that coordinates transcriptional and post-transcriptional repression of hTERT during cellular differentiation. Deregulation of HOXC5 and mir-615-3p expression may contribute to the activation of hTERT in human cancers.


Assuntos
Diferenciação Celular/genética , Transformação Celular Neoplásica/genética , Proteínas de Homeodomínio/genética , MicroRNAs/genética , Telomerase/biossíntese , Regiões 3' não Traduzidas/genética , Regiões 5' não Traduzidas/genética , Animais , Linhagem Celular Tumoral , Elementos Facilitadores Genéticos/genética , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Células MCF-7 , Camundongos , Neoplasias/genética , Neoplasias/patologia , Regiões Promotoras Genéticas/genética
14.
Cancer Cell ; 32(5): 639-653.e6, 2017 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-29136507

RESUMO

Cancers grow within tissues through molecular mechanisms still unclear. Invasiveness correlates with perturbed O-glycosylation, a covalent modification of cell-surface proteins. Here, we show that, in human and mouse liver cancers, initiation of O-glycosylation by the GALNT glycosyl-transferases increases and shifts from the Golgi to the endoplasmic reticulum (ER). In a mouse liver cancer model, expressing an ER-targeted GALNT1 (ER-G1) massively increased tumor expansion, with median survival reduced from 23 to 10 weeks. In vitro cell growth was unaffected, but ER-G1 strongly enabled matrix degradation and tissue invasion. Unlike its Golgi-localized counterpart, ER-G1 glycosylates the matrix metalloproteinase MMP14, a process required for tumor expansion. Together, our results indicate that GALNTs strongly promote liver tumor growth after relocating to the ER.


Assuntos
Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Neoplasias Hepáticas/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Animais , Western Blotting , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Glicosilação , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Metaloproteinase 14 da Matriz/genética , Camundongos Endogâmicos C57BL , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , Metástase Neoplásica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Polipeptídeo N-Acetilgalactosaminiltransferase
15.
Mol Biol Cell ; 28(25): 3686-3698, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29021342

RESUMO

The synthesis of glycans and the sorting of proteins are critical functions of the Golgi apparatus and depend on its highly complex and compartmentalized architecture. High-content image analysis coupled to RNA interference screening offers opportunities to explore this organelle organization and the gene network underlying it. To date, image-based Golgi screens have based on a single parameter or supervised analysis with predefined Golgi structural classes. Here, we report the use of multiparametric data extracted from a single marker and a computational unsupervised analysis framework to explore Golgi phenotypic diversity more extensively. In contrast with the three visually definable phenotypes, our framework reproducibly identified 10 Golgi phenotypes. They were used to quantify and stratify phenotypic similarities among genetic perturbations. The derived phenotypic network partially overlaps previously reported protein-protein interactions as well as suggesting novel functional interactions. Our workflow suggests the existence of multiple stable Golgi organizational states and provides a proof of concept for the classification of drugs and genes using fine-grained phenotypic information.


Assuntos
Complexo de Golgi/fisiologia , Aprendizado de Máquina não Supervisionado , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Células HeLa , Ensaios de Triagem em Larga Escala/métodos , Humanos , Fenótipo , Polissacarídeos/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Reprodutibilidade dos Testes
17.
PLoS Genet ; 13(4): e1006698, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28403141

RESUMO

The cellular machinery required for the fusion of constitutive secretory vesicles with the plasma membrane in metazoans remains poorly defined. To address this problem we have developed a powerful, quantitative assay for measuring secretion and used it in combination with combinatorial gene depletion studies in Drosophila cells. This has allowed us to identify at least three SNARE complexes mediating Golgi to PM transport (STX1, SNAP24/29 and Syb; STX1, SNAP24/29 and YKT6; STX4, SNAP24 and Syb). RNAi mediated depletion of YKT6 and VAMP3 in mammalian cells also blocks constitutive secretion suggesting that YKT6 has an evolutionarily conserved role in this process. The unexpected role of YKT6 in plasma membrane fusion may in part explain why RNAi and gene disruption studies have failed to produce the expected phenotypes in higher eukaryotes.


Assuntos
Membrana Celular/genética , Proteínas de Drosophila/genética , Proteínas R-SNARE/genética , Proteínas SNARE/genética , Proteína 3 Associada à Membrana da Vesícula/genética , Animais , Membrana Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Heterozigoto , Humanos , Fusão de Membrana/genética , Transporte Proteico/genética , Proteínas R-SNARE/metabolismo , Interferência de RNA , Proteínas SNARE/metabolismo , Toxina Shiga I/genética , Toxina Shiga I/metabolismo , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/genética , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo , Proteína 3 Associada à Membrana da Vesícula/metabolismo
19.
Nat Commun ; 7: 13150, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27748395

RESUMO

Enterovirus 71 (EV71) is a neurotropic enterovirus without antivirals or vaccine, and its host-pathogen interactions remain poorly understood. Here we use a human genome-wide RNAi screen to identify 256 host factors involved in EV71 replication in human rhabdomyosarcoma cells. Enrichment analyses reveal overrepresentation in processes like mitotic cell cycle and transcriptional regulation. We have carried out orthogonal experiments to characterize the roles of selected factors involved in cell cycle regulation and endoplasmatic reticulum-associated degradation. We demonstrate nuclear egress of CDK6 in EV71 infected cells, and identify CDK6 and AURKB as resistance factors. NGLY1, which co-localizes with EV71 replication complexes at the endoplasmatic reticulum, supports EV71 replication. We confirm importance of these factors for EV71 replication in a human neuronal cell line and for coxsackievirus A16 infection. A small molecule inhibitor of NGLY1 reduces EV71 replication. This study provides a comprehensive map of EV71 host factors and reveals potential antiviral targets.


Assuntos
Enterovirus Humano A/crescimento & desenvolvimento , Genoma Humano/genética , Interferência de RNA , Replicação Viral , Linhagem Celular Tumoral , Resistência à Doença/genética , Enterovirus Humano A/fisiologia , Regulação Neoplásica da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Rabdomiossarcoma/genética , Rabdomiossarcoma/patologia , Rabdomiossarcoma/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...