Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Cell Biol ; 218(2): 422-432, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30602538

RESUMO

53BP1 is a chromatin-associated protein that regulates the DNA damage response. In this study, we identify the TPX2/Aurora A heterodimer, nominally considered a mitotic kinase complex, as a novel binding partner of 53BP1. We find that TPX2/Aurora A plays a previously unrecognized role in DNA damage repair and replication fork stability by counteracting 53BP1 function. Loss of TPX2 or Aurora A compromises DNA end resection, BRCA1 and Rad51 recruitment, and homologous recombination. Furthermore, loss of TPX2 or Aurora A causes deprotection of stalled replication forks upon replication stress induction. This fork protection pathway counteracts MRE11 nuclease activity but functions in parallel to BRCA1. Strikingly, concurrent loss of 53BP1 rescues not only BRCA1/Rad51 recruitment but also the fork instability induced upon TPX2 loss. Our work suggests the presence of a feedback mechanism by which 53BP1 is regulated by a novel binding partner and uncovers a unique role for 53BP1 in replication fork stability.


Assuntos
Aurora Quinase A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Replicação do DNA , Recombinação Homóloga , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Animais , Aurora Quinase A/genética , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/genética , Células HeLa , Humanos , Proteína Homóloga a MRE11/genética , Proteína Homóloga a MRE11/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética
2.
Cell Rep ; 18(2): 432-442, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28076787

RESUMO

Natural products have demonstrated utility in the clinic and can also act as probes to understand complex cellular pathways. Sanglifehrin A (SFA) is a mixed polyketide and non-ribosomal peptide synthase natural product with sub-nano-molar affinity for its receptor cyclophilin A (PPIA). It has been shown to behave in vitro as an immune suppressant. Here, we identify inosine-5'-monophosphate dehydrogenase 2 (IMPDH2) as an intracellular target of the PPIA-SFA binary complex. The formation of this ternary complex does not inhibit the enzymatic activity of IMPDH2. Rather, ternary complex formation modulates cell growth through interaction with the cystathionine-ß-synthase (CBS) domain of IMPDH2. We further demonstrate that the SFA complex is highly isoform selective for IMPDH2 (versus IMPDH1). This work reveals a role for the CBS domains of IMPDH2 in cellular proliferation, suggesting a more complex role than previously suspected for IMPDH2 in T cell activation and proliferation.


Assuntos
Ciclofilina A/metabolismo , IMP Desidrogenase/metabolismo , Proliferação de Células , Humanos , IMP Desidrogenase/química , Células Jurkat , Células K562 , Lactonas/química , Lactonas/metabolismo , Ligação Proteica , Domínios Proteicos , Compostos de Espiro/química , Compostos de Espiro/metabolismo , Relação Estrutura-Atividade
3.
Proc Natl Acad Sci U S A ; 111(52): 18631-6, 2014 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-25512513

RESUMO

The signal transduction pathway of the DNA damage response (DDR) is activated to maintain genomic integrity following DNA damage. The DDR promotes genomic integrity by regulating a large network of cellular activities that range from DNA replication and repair to transcription, RNA splicing, and metabolism. In this study we define an interaction between the DDR factor NBS1 and TCOF1, a nucleolar protein that regulates ribosomal DNA (rDNA) transcription and is mutated in Treacher Collins syndrome. We show that NBS1 relocalizes to nucleoli after DNA damage in a manner dependent on TCOF1 and on casein kinase II and ATM, which are known to modify TCOF1 by phosphorylation. Moreover, we identify a putative ATM phosphorylation site that is required for NBS1 relocalization to nucleoli in response to DNA damage. Last, we report that TCOF1 promotes cellular resistance to DNA damaging agents. Collectively, our findings identify TCOF1 as a DDR factor that could cooperate with ATM and NBS1 to suppress inappropriate rDNA transcription and maintain genomic integrity after DNA damage.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Nucléolo Celular/metabolismo , Dano ao DNA , DNA Ribossômico/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Transporte Ativo do Núcleo Celular/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Nucléolo Celular/genética , DNA Ribossômico/genética , Humanos , Proteínas Nucleares/genética , Fosfoproteínas/genética , Fosforilação/genética , Transcrição Gênica/genética
4.
J Cell Biol ; 203(3): 457-70, 2013 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-24217620

RESUMO

Histone demethylation is known to regulate transcription, but its role in other processes is largely unknown. We report a role for the histone demethylase LSD1/KDM1A in the DNA damage response (DDR). We show that LSD1 is recruited directly to sites of DNA damage. H3K4 dimethylation, a major substrate for LSD1, is reduced at sites of DNA damage in an LSD1-dependent manner. The E3 ubiquitin ligase RNF168 physically interacts with LSD1 and we find this interaction to be important for LSD1 recruitment to DNA damage sites. Although loss of LSD1 did not affect the initial formation of pH2A.X foci, 53BP1 and BRCA1 complex recruitment were reduced upon LSD1 knockdown. Mechanistically, this was likely a result of compromised histone ubiquitylation preferentially in late S/G2. Consistent with a role in the DDR, knockdown of LSD1 resulted in moderate hypersensitivity to γ-irradiation and increased homologous recombination. Our findings uncover a direct role for LSD1 in the DDR and place LSD1 downstream of RNF168 in the DDR pathway.


Assuntos
Reparo do DNA/genética , Histona Desmetilases/metabolismo , Histonas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Proteína BRCA1/metabolismo , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Dano ao DNA , Metilação de DNA , Células HEK293 , Células HeLa , Histona Desmetilases/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Interferência de RNA , RNA Interferente Pequeno , Tolerância a Radiação , Fase S/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53 , Ubiquitinação
5.
Proc Natl Acad Sci U S A ; 109(23): E1473-80, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22547818

RESUMO

The Notch signaling pathway is a key determinant in keratinocyte differentiation and growth cycle arrest, and has been reported to have a tumor suppressor function in skin. The papillomavirus life cycle is intricately linked to the differentiation status of keratinocytes. Papillomaviruses are associated with benign proliferative epithelial lesions in their respective hosts. Although human papillomaviruses (HPVs) associated with genital tract lesions have been extensively studied, studies of the cutaneous HPVs are more limited. In particular, it is well established that the E6 proteins of high-risk HPVs of the α-genus such as HPV16 and HPV18 mediate the degradation of p53 by its association with the ubiquitin ligase E6AP. In contrast, less is known about the cellular activities of the cutaneous HPVs of the ß-genus. By using an unbiased proteomic approach, we identify MAML1 and other members of the Notch transcription complex as high-confidence cellular interacting proteins of E6 proteins of the ß-genus HPVs and of the bovine papillomavirus type 1 associated with cutaneous fibropapillomas. We show that bovine papillomavirus type 1 and ß-HPV E6 repress Notch transcriptional activation, and that this repression is dependent on an interaction with MAML1. Finally, we show that the expression levels of endogenous Notch target genes are repressed by ß-HPV E6 proteins. These findings elucidate a mechanism of viral antagonism of Notch signaling, and suggest that Notch signaling is an important epithelial cell pathway target for the ß-HPVs.


Assuntos
Betapapillomavirus/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Animais , Western Blotting , Linhagem Celular , Humanos , Luciferases , Camundongos , Plasmídeos/genética , Proteômica , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética , Transfecção
6.
Mol Cell Biol ; 32(15): 3095-106, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22645313

RESUMO

The E6AP ubiquitin ligase catalyzes the high-risk human papillomaviruses' E6-mediated ubiquitylation of p53, contributing to the neoplastic progression of cells infected by these viruses. Defects in the activity and the dosage of E6AP are linked to Angelman syndrome and to autism spectrum disorders, respectively, highlighting the need for precise control of the enzyme. With the exception of HERC2, which modulates the ubiquitin ligase activity of E6AP, little is known about the regulation or function of E6AP normally. Using a proteomic approach, we have identified and validated several new E6AP-interacting proteins, including HIF1AN, NEURL4, and mitogen-activated protein kinase 6 (MAPK6). E6AP exists as part of several different protein complexes, including the proteasome and an independent high-molecular-weight complex containing HERC2, NEURL4, and MAPK6. In examining the functional consequence of its interaction with the proteasome, we found that UBE3C (another proteasome-associated ubiquitin ligase), but not E6AP, contributes to proteasomal processivity in mammalian cells. We also found that E6 associates with the HERC2-containing high-molecular-weight complex through its binding to E6AP. These proteomic studies reveal a level of complexity for E6AP that has not been previously appreciated and identify a number of new cellular proteins through which E6AP may be regulated or functioning.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Oxigenases de Função Mista/metabolismo , Proteínas Repressoras/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Síndrome de Angelman/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Células HeLa , Humanos , Proteína Quinase 6 Ativada por Mitógeno/genética , Oxigenases de Função Mista/genética , Papillomaviridae/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteômica , Interferência de RNA , RNA Interferente Pequeno , Proteínas Repressoras/genética , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
7.
Proc Natl Acad Sci U S A ; 109(13): 4828-33, 2012 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-22411829

RESUMO

UHRF1 (Ubiquitin-like, with PHD and RING finger domains 1) plays an important role in DNA CpG methylation, heterochromatin function and gene expression. Overexpression of UHRF1 has been suggested to contribute to tumorigenesis. However, regulation of UHRF1 is largely unknown. Here we show that the deubiquitylase USP7 interacts with UHRF1. Using interaction-defective and catalytic mutants of USP7 for complementation experiments, we demonstrate that both physical interaction and catalytic activity of USP7 are necessary for UHRF1 ubiquitylation and stability regulation. Mass spectrometry analysis identified phosphorylation of serine (S) 652 within the USP7-interacting domain of UHRF1, which was further confirmed by a UHRF1 S652 phosphor (S652ph)-specific antibody. Importantly, the S652ph antibody identifies phosphorylated UHRF1 in mitotic cells and consistently S652 can be phosphorylated by the M phase-specific kinase CDK1-cyclin B in vitro. UHRF1 S652 phosphorylation significantly reduces UHRF1 interaction with USP7 in vitro and in vivo, which is correlated with a decreased UHRF1 stability in the M phase of the cell cycle. In contrast, UHRF1 carrying the S652A mutation, which renders UHRF1 resistant to phosphorylation at S652, is more stable. Importantly, cells carrying the S652A mutant grow more slowly suggesting that maintaining an appropriate level of UHRF1 is important for cell proliferation regulation. Taken together, our findings uncovered a cell cycle-specific signaling event that relieves UHRF1 from its interaction with USP7, thus exposing UHRF1 to proteasome-mediated degradation. These findings identify a molecular mechanism by which cellular UHRF1 level is regulated, which may impact cell proliferation.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Divisão Celular , Epigênese Genética , Ubiquitina Tiolesterase/metabolismo , Sequência de Aminoácidos , Aminoácidos/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/química , Proteína Quinase CDC2/metabolismo , Linhagem Celular , Cromatografia de Afinidade , Ciclina B/metabolismo , Estabilidade Enzimática , Humanos , Dados de Sequência Molecular , Fosforilação , Fosfosserina/metabolismo , Estrutura Terciária de Proteína , Ubiquitina Tiolesterase/química , Ubiquitina-Proteína Ligases , Peptidase 7 Específica de Ubiquitina , Ubiquitinação
8.
Proc Natl Acad Sci U S A ; 109(5): E260-7, 2012 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-22232672

RESUMO

More than 120 human papillomaviruses (HPVs) have now been identified and have been associated with a variety of clinical lesions. To understand the molecular differences among these viruses that result in lesions with distinct pathologies, we have begun a MS-based proteomic analysis of HPV-host cellular protein interactions and have created the plasmid and cell line libraries required for these studies. To validate our system, we have characterized the host cellular proteins that bind to the E7 proteins expressed from 17 different HPV types. These studies reveal a number of interactions, some of which are conserved across HPV types and others that are unique to a single HPV species or HPV genus. Binding of E7 to UBR4/p600 is conserved across all virus types, whereas the cellular protein ENC1 binds specifically to the E7s from HPV18 and HPV45, both members of genus alpha, species 7. We identify a specific interaction of HPV16 E7 with ZER1, a substrate specificity factor for a cullin 2 (CUL2)-RING ubiquitin ligase, and show that ZER1 is required for the binding of HPV16 E7 to CUL2. We further show that ZER1 is required for the destabilization of the retinoblastoma tumor suppressor RB1 in HPV16 E7-expressing cells and propose that a CUL2-ZER1 complex functions to target RB1 for degradation in HPV16 E7-expressing cells. These studies refine the current understanding of HPV E7 functions and establish a platform for the rapid identification of virus-host interactions.


Assuntos
Proteínas Oncogênicas Virais/metabolismo , Papillomaviridae/metabolismo , Sequência de Aminoácidos , Western Blotting , Linhagem Celular , Humanos , Dados de Sequência Molecular , Proteínas Oncogênicas Virais/química , Papillomaviridae/classificação , Ligação Proteica , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Espectrometria de Massas em Tandem
9.
Nat Cell Biol ; 14(1): 93-105, 2011 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-22119785

RESUMO

Proteins that fail to correctly fold or assemble into oligomeric complexes in the endoplasmic reticulum (ER) are degraded by a ubiquitin- and proteasome-dependent process known as ER-associated degradation (ERAD). Although many individual components of the ERAD system have been identified, how these proteins are organized into a functional network that coordinates recognition, ubiquitylation and dislocation of substrates across the ER membrane is not well understood. We have investigated the functional organization of the mammalian ERAD system using a systems-level strategy that integrates proteomics, functional genomics and the transcriptional response to ER stress. This analysis supports an adaptive organization for the mammalian ERAD machinery and reveals a number of metazoan-specific genes not previously linked to ERAD.


Assuntos
Degradação Associada com o Retículo Endoplasmático/fisiologia , Retículo Endoplasmático/metabolismo , Sequência de Aminoácidos , Animais , Células HEK293 , Células HeLa , Humanos , Dados de Sequência Molecular , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Proteínas/metabolismo , Proteólise , Interferência de RNA , Receptores do Fator Autócrino de Motilidade , Ubiquitina-Proteína Ligases/metabolismo
10.
Mol Cell ; 44(3): 373-84, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-22055184

RESUMO

Demethylation by the AlkB dioxygenases represents an important mechanism for repair of N-alkylated nucleotides. However, little is known about their functions in mammalian cells. We report the purification of the ALKBH3 complex and demonstrate its association with the activating signal cointegrator complex (ASCC). ALKBH3 is overexpressed in various cancers, and both ALKBH3 and ASCC are important for alkylation damage resistance in these tumor cell lines. ASCC3, the largest subunit of ASCC, encodes a 3'-5' DNA helicase, whose activity is crucial for the generation of single-stranded DNA upon which ALKBH3 preferentially functions for dealkylation. In cell lines that are dependent on ALKBH3 and ASCC3 for alkylation damage resistance, loss of ALKBH3 or ASCC3 leads to increased 3-methylcytosine and reduced cell proliferation, which correlates with pH2A.X and 53BP1 foci formation. Our data provide a molecular mechanism by which ALKBH3 collaborates with ASCC to maintain genomic integrity in a cell-type specific manner.


Assuntos
Proliferação de Células , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA , Dioxigenases/metabolismo , Neoplasias da Próstata/enzimologia , Homólogo AlkB 3 da Dioxigenase Dependente de alfa-Cetoglutarato , Alquilação , Animais , Antineoplásicos Alquilantes/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA Helicases/genética , Enzimas Reparadoras do DNA/genética , Dioxigenases/genética , Relação Dose-Resposta a Droga , Células HEK293 , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Metanossulfonato de Metila , Camundongos , Camundongos Endogâmicos NOD , Mutação , Transplante de Neoplasias , Fosforilação , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Interferência de RNA , Fatores de Tempo , Transfecção , Carga Tumoral , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
11.
Science ; 332(6035): 1313-7, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21659603

RESUMO

The DNA damage response (DDR) is brought about by a protein kinase cascade that orchestrates DNA repair through transcriptional and posttranslational mechanisms. Cell cycle arrest is a hallmark of the DDR. We screened for cells that lacked damage-induced cell cycle arrest and uncovered a critical role for Fanconi anemia and homologous recombination proteins in ATR (ataxia telangiectasia and Rad3-related) signaling. Three DDR candidates, the RNA processing protein INTS7, the circadian transcription factor CLOCK, and a previously uncharacterized protein RHINO, were recruited to sites of DNA damage. RHINO independently bound the Rad9-Rad1-Hus1 complex (9-1-1) and the ATR activator TopBP1. RHINO was recruited to sites of DNA damage by the 9-1-1 complex to promote Chk1 activation. We suggest that RHINO functions together with the 9-1-1 complex and TopBP1 to fully activate ATR.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas de Ciclo Celular/metabolismo , Quimiocinas/fisiologia , Reparo do DNA , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Transporte/metabolismo , Ciclo Celular/genética , Linhagem Celular Tumoral , Quimiocinas/genética , Quimiocinas CXC , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Exonucleases/metabolismo , Humanos , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo
12.
Mol Cell Biol ; 31(13): 2641-52, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21555454

RESUMO

Bromodomain protein 4 (Brd4) plays critical roles in development, cancer progression, and virus-host pathogenesis. To gain mechanistic insight into the various biological functions of Brd4, we performed a proteomic analysis to identify and characterize Brd4-associated cellular proteins. We found that the extraterminal (ET) domain, whose function has to date not been determined, interacts with NSD3, JMJD6, CHD4, GLTSCR1, and ATAD5. These ET-domain interactions were also conserved for Brd2 and Brd3, the other human BET proteins tested. We demonstrated that GLTSCR1, NSD3, and JMJD6 impart a pTEFb-independent transcriptional activation function on Brd4. NSD3 as well as JMJD6 is recruited to regulated genes in a Brd4-dependent manner. Moreover, we found that depletion of Brd4 or NSD3 reduces H3K36 methylation, demonstrating that the Brd4/NSD3 complex regulates this specific histone modification. Our results indicate that the Brd4 ET domain through the recruitment of the specific effectors regulates transcriptional activity. In particular, we show that one of these effectors, NSD3, regulates transcription by modifying the chromatin microenvironment at Brd4 target genes. Our study thus identifies the ET domain as a second important transcriptional regulatory domain for Brd4 in addition to the carboxyl-terminal domain (CTD) that interacts with pTEFb.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Proteínas Nucleares/metabolismo , Fator B de Elongação Transcricional Positiva/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Proteínas de Ciclo Celular , Linhagem Celular , Proteínas Cromossômicas não Histona , Humanos , Histona Desmetilases com o Domínio Jumonji , Dados de Sequência Molecular , Proteínas Nucleares/genética , Estrutura Terciária de Proteína/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/metabolismo
13.
Mol Cell ; 40(4): 645-57, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21055985

RESUMO

Replication stress involving collision of replisomes with camptothecin (CPT)-stabilized DNA-Topoisomerase I adducts activates an ATR-dependent pathway to promote repair by homologous recombination. To identify human genes that protect cells from such replication stress, we performed a genome-wide CPT sensitivity screen. Among numerous candidate genes are two previously unstudied proteins: the ankyrin repeat protein NFKBIL2 and C6ORF167 (MMS22L), distantly related to yeast replication stress regulator Mms22p. MMS22L and NFKBIL2 interact with each other and with FACT (facilitator of chromatin transcription) and MCM (minichromosome maintenance) complexes. Cells depleted of NFKBIL2 or MMS22L are sensitive to DNA-damaging agents, load phosphorylated RPA onto chromatin in a CTIP-dependent manner, activate the ATR/ATRIP-CHK1 and double-strand break repair signaling pathways, and are defective in HR. This study identifies MMS22L-NFKBIL2 as components of the replication stress control pathway and provides a resource for discovery of additional components of this pathway.


Assuntos
Camptotecina/farmacologia , Proteínas de Ligação a DNA/metabolismo , Testes Genéticos , Genoma Humano/genética , Instabilidade Genômica/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , DNA Polimerase Dirigida por DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Células HeLa , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Complexos Multienzimáticos/metabolismo , NF-kappa B/deficiência , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Recombinação Genética/efeitos dos fármacos , Recombinação Genética/genética , Proteína de Replicação A/metabolismo , Reprodutibilidade dos Testes , Estresse Fisiológico/efeitos dos fármacos , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
14.
J Virol ; 84(9): 4451-60, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20181716

RESUMO

The papillomavirus E2 open reading frame encodes the full-length E2 protein as well as an alternatively spliced product called E8;E2C. E8;E2C has been best studied for the high-risk human papillomaviruses, where it has been shown to regulate viral genome levels and, like the full-length E2 protein, to repress transcription from the viral promoter that directs the expression of the viral E6 and E7 oncogenes. The repression function of E8;E2C is dependent on the 12-amino-acid N-terminal sequence from the E8 open reading frame (ORF). In order to understand the mechanism by which E8;E2C mediates transcriptional repression, we performed an unbiased proteomic analysis from which we identified six high-confidence candidate interacting proteins (HCIPs) for E8;E2C; the top two are NCoR1 and TBLR1. We established an interaction of E8;E2C with an NCoR1/HDAC3 complex and demonstrated that this interaction requires the wild-type E8 open reading frame. Small interfering RNA (siRNA) knockdown studies demonstrated the involvement of NCoR1/HDAC3 in the E8;E2C-dependent repression of the viral long control region (LCR) promoter. Additional genetic work confirmed that the papillomavirus E2 and E8;E2C proteins repress transcription through distinct mechanisms.


Assuntos
Regulação Viral da Expressão Gênica , Inativação Gênica , Correpressor 1 de Receptor Nuclear/metabolismo , Papillomaviridae/fisiologia , Proteínas Repressoras/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Técnicas de Silenciamento de Genes , Humanos , Modelos Biológicos , Ligação Proteica , Mapeamento de Interação de Proteínas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
15.
Proc Natl Acad Sci U S A ; 107(8): 3752-7, 2010 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-20133580

RESUMO

An essential step in the pathogenesis of human papillomavirus (HPV)-associated cancers is the dysregulated expression of the viral oncogenes. The papillomavirus E2 protein can silence the long control region (LCR) promoter that controls viral E6 and E7 oncogene expression. The mechanisms by which E2 represses oncogene expression and the cellular factors through which E2 mediates this silencing are largely unknown. We conducted an unbiased, genome-wide siRNA screen and series of secondary screens that identified 96 cellular genes that contribute to the repression of the HPV LCR. In addition to confirming a role for the E2-binding bromodomain protein Brd4 in E2-mediated silencing, we identified a number of genes that have not previously been implicated in E2 repression, including the demethylase JARID1C/SMCX as well as EP400, a component of the NuA4/TIP60 histone acetyltransferase complex. Each of these genes contributes independently and additively to E2-mediated silencing, indicating that E2 functions through several distinct cellular complexes to repress E6 and E7 expression.


Assuntos
DNA Helicases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Viral da Expressão Gênica , Papillomavirus Humano 18/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas Virais/genética , Oxirredutases N-Desmetilantes/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , DNA Helicases/genética , Feminino , Inativação Gênica , Estudo de Associação Genômica Ampla , Células HeLa , Histona Desmetilases , Humanos , Proteínas Nucleares/genética , Proteínas Oncogênicas Virais/metabolismo , Oxirredutases N-Desmetilantes/genética , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/virologia , RNA Interferente Pequeno/genética , Fatores de Transcrição/genética , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/virologia , Proteínas Virais/metabolismo , Replicação Viral/genética
16.
Genes Dev ; 23(20): 2415-25, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19793862

RESUMO

The integrity of genomic DNA is continuously challenged by the presence of DNA base lesions or DNA strand breaks. Here we report the identification of a new DNA damage response protein, SMARCAL1 (SWI/SNF-related, matrix associated, actin-dependent regulator of chromatin, subfamily a-like 1), which is a member of the SNF2 family and is mutated in Schimke immunoosseous dysplasia (SIOD). We demonstrate that SMARCAL1 directly interacts with Replication protein A (RPA) and is recruited to sites of DNA damage in an RPA-dependent manner. SMARCAL1-depleted cells display sensitivity to DNA-damaging agents that induce replication fork collapse, and exhibit slower fork recovery and delayed entry into mitosis following S-phase arrest. Furthermore, SIOD patient fibroblasts reconstituted with SMARCAL1 exhibit faster cell cycle progression after S-phase arrest. Thus, the symptoms of SIOD may be caused, at least in part, by defects in the cellular response to DNA replication stress.


Assuntos
DNA Helicases/metabolismo , Osteocondrodisplasias/fisiopatologia , Proteína de Replicação A/metabolismo , Sequência de Aminoácidos , Ciclo Celular , Linhagem Celular , Dano ao DNA , DNA Helicases/química , Replicação do DNA , Humanos , Dados de Sequência Molecular , Osteocondrodisplasias/genética , Alinhamento de Sequência
17.
Mol Biol Cell ; 19(12): 5059-71, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18799622

RESUMO

Fused Toes (FTS) is a member of a small group of inactive variant E2 ubiquitin-conjugating enzyme domain-containing proteins of unknown function. Through proteomic analysis of FTS complexes purified from human embryonic kidney 293T cells, we identified a new multiprotein complex, the FHF complex, containing FTS, members of the microtubule-binding Hook family of coiled-coil proteins (Hook1, Hook2, and Hook3), and a previously uncharacterized 107-kDa protein, FTS and Hook Interacting Protein (FHIP). FTS associated with a conserved C-terminal motif in Hook proteins in the yeast two-hybrid system and in tissue culture cells, and Hook proteins were found to form homo- and heterodimers. The approximately 500-kDa FHF complex contained all three Hook proteins, and small interfering RNA depletion experiments suggest that Hook proteins can interact interchangeably within this complex. Hook proteins as well as FTS interact with members of both the class B and class C components of the homotypic vesicular protein sorting (HOPS) complex. Depletion of FTS by RNA interference affects both the trafficking of epidermal growth factor from early-to-late endosome/lysosomes and the efficiency by which overexpression of the HOPS component Vps18 promotes clustering of lysosomal-associated membrane protein 1-positive endosome/lysosomes. These data suggest that the FTS/Hook/FHIP complex functions to promote vesicle trafficking and/or fusion via the HOPS complex.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Endossomos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Complexos Multiproteicos/metabolismo , Proteína p107 Retinoblastoma-Like/metabolismo , Vacúolos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular , Humanos , Lisossomos/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Interferência de RNA , Proteína p107 Retinoblastoma-Like/genética , Alinhamento de Sequência , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo
18.
Science ; 319(5863): 620-4, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18239126

RESUMO

Retroviral short hairpin RNA (shRNA)-mediated genetic screens in mammalian cells are powerful tools for discovering loss-of-function phenotypes. We describe a highly parallel multiplex methodology for screening large pools of shRNAs using half-hairpin barcodes for microarray deconvolution. We carried out dropout screens for shRNAs that affect cell proliferation and viability in cancer cells and normal cells. We identified many shRNAs to be antiproliferative that target core cellular processes, such as the cell cycle and protein translation, in all cells examined. Moreover, we identified genes that are selectively required for proliferation and survival in different cell lines. Our platform enables rapid and cost-effective genome-wide screens to identify cancer proliferation and survival genes for target discovery. Such efforts are complementary to the Cancer Genome Atlas and provide an alternative functional view of cancer cells.


Assuntos
Neoplasias da Mama/genética , Proliferação de Células , Neoplasias do Colo/genética , Genes Neoplásicos , Genômica/métodos , Neoplasias da Mama/patologia , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Neoplasias do Colo/patologia , Biblioteca Gênica , Vetores Genéticos , Genoma Humano , Humanos , MicroRNAs , Análise de Sequência com Séries de Oligonucleotídeos , RNA Interferente Pequeno , Retroviridae/genética
19.
Proc Natl Acad Sci U S A ; 104(21): 8869-74, 2007 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-17495026

RESUMO

Mutations in the cylindromatosis (CYLD) gene cause benign tumors of skin appendages, referred to as cylindromas. The CYLD gene encodes a deubiquitinating enzyme that removes Lys-63-linked ubiquitin chains from I kappa B kinase signaling components and thereby inhibits NF-kappaB pathway activation. The dysregulation of NF-kappaB activity has been proposed to promote cell transformation in part by increasing apoptosis resistance, but it is not clear whether this is CYLD's only or predominant tumor-suppressing function. Here, we show that CYLD is also required for timely entry into mitosis. Consistent with a cell-cycle regulatory function, CYLD localizes to microtubules in interphase and the midbody during telophase, and its protein levels decrease as cells exit from mitosis. We identified the protein kinase Plk1 as a potential target of CYLD in the regulation of mitotic entry, based on their physical interaction and similar loss-of-function and overexpression phenotypes. Our findings raise the possibility that, as with other genes regulating tumorigenesis, CYLD has not only tumor-suppressing (apoptosis regulation) but also tumor-promoting activities (enhancer of mitotic entry). We propose that this additional function of CYLD could provide an explanation for the benign nature of most cylindroma lesions.


Assuntos
Mitose , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Enzima Desubiquitinante CYLD , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , NF-kappa B/metabolismo , Ligação Proteica , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteínas Elk-1 do Domínio ets/química , Proteínas Elk-1 do Domínio ets/genética , Proteínas Elk-1 do Domínio ets/metabolismo
20.
Mol Cell ; 26(1): 131-43, 2007 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-17434132

RESUMO

The ubiquitin-mediated proteolysis of cyclin E plays a central role in cell-cycle progression, and cyclin E accumulation is a common event in cancer. Cyclin E degradation is triggered by multisite phosphorylation, which induces binding to the SCF(Fbw7) ubiquitin ligase complex. Structures of the Skp1-Fbw7 complex bound to cyclin E peptides identify a doubly phosphorylated pThr380/pSer384 cyclin E motif as an optimal, high-affinity degron and a singly phosphorylated pThr62 motif as a low-affinity one. Biochemical data indicate that the closely related yeast SCF(Cdc4) complex recognizes the multisite phosphorylated Sic1 substrate similarly and identify three doubly phosphorylated Sic1 degrons, each capable of high-affinity interactions with two Cdc4 phosphate binding sites. A model that explains the role of multiple cyclin E/Sic1 degrons is provided by the findings that Fbw7 and Cdc4 dimerize, that Fbw7 dimerization enhances the turnover of a weakly associated cyclin E in vivo, and that Cdc4 dimerization increases the rate and processivity of Sic1 ubiquitination in vitro.


Assuntos
Proteínas de Ciclo Celular/química , Ciclina E/química , Proteínas F-Box/química , Proteínas Quinases Associadas a Fase S/química , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Proteínas Inibidoras de Quinase Dependente de Ciclina , Dimerização , Proteínas F-Box/isolamento & purificação , Proteínas F-Box/metabolismo , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Fosfopeptídeos/química , Fosfopeptídeos/metabolismo , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Quinases Associadas a Fase S/isolamento & purificação , Proteínas Quinases Associadas a Fase S/metabolismo , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Homologia de Sequência de Aminoácidos , Serina/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato , Treonina/metabolismo , Transfecção , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA