Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Cell ; 84(2): 386-400.e11, 2024 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-38103558

RESUMO

The posttranslational modifier ubiquitin regulates most cellular processes. Its ability to form polymeric chains of distinct linkages is key to its diverse functionality. Yet, we still lack the experimental tools to induce linkage-specific polyubiquitylation of a protein of interest in cells. Here, we introduce a set of engineered ubiquitin protein ligases and matching ubiquitin acceptor tags for the rapid, inducible linear (M1-), K48-, or K63-linked polyubiquitylation of proteins in yeast and mammalian cells. By applying the so-called "Ubiquiton" system to proteasomal targeting and the endocytic pathway, we validate this tool for soluble cytoplasmic and nuclear as well as chromatin-associated and integral membrane proteins and demonstrate how it can be used to control the localization and stability of its targets. We expect that the Ubiquiton system will serve as a versatile, broadly applicable research tool to explore the signaling functions of polyubiquitin chains in many biological contexts.


Assuntos
Ubiquitina-Proteína Ligases , Ubiquitina , Animais , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Poliubiquitina/genética , Poliubiquitina/metabolismo , Transdução de Sinais , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitinação , Mamíferos/metabolismo
2.
Mol Cell ; 77(1): 3-16.e4, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31607544

RESUMO

Tracing DNA repair factors by fluorescence microscopy provides valuable information about how DNA damage processing is orchestrated within cells. Most repair pathways involve single-stranded DNA (ssDNA), making replication protein A (RPA) a hallmark of DNA damage and replication stress. RPA foci emerging during S phase in response to tolerable loads of polymerase-blocking lesions are generally thought to indicate stalled replication intermediates. We now report that in budding yeast they predominantly form far away from sites of ongoing replication, and they do not overlap with any of the repair centers associated with collapsed replication forks or double-strand breaks. Instead, they represent sites of postreplicative DNA damage bypass involving translesion synthesis and homologous recombination. We propose that most RPA and recombination foci induced by polymerase-blocking lesions in the replication template are clusters of repair tracts arising from replication centers by polymerase re-priming and subsequent expansion of daughter-strand gaps over the course of S phase.


Assuntos
Replicação do DNA/genética , DNA de Cadeia Simples/genética , DNA Polimerase Dirigida por DNA/genética , Genoma/genética , Dano ao DNA/genética , Reparo do DNA/genética , Recombinação Homóloga/genética , Proteína de Replicação A/genética , Fase S/genética , Saccharomycetales/genética
3.
J Am Chem Soc ; 146(11): 7135-7139, 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38441879

RESUMO

Organic near-infrared (NIR) photoblinking fluorophores are highly desirable for live-cell super-resolution imaging based on single-molecule localization microscopy (SMLM). Herein we introduce a novel small chromophore, PMIP, through the fusion of perylenecarboximide with 2,2-dimetheylpyrimidine. PMIP exhibits an emission maximum at 732 nm with a high fluorescence quantum yield of 60% in the wavelength range of 700-1000 nm and excellent photoblinking without any additives. With resorcinol-functionalized PMIP (PMIP-OH), NIR SMLM imaging of lysosomes is demonstrated for the first time in living mammalian cells under physiological conditions. Moreover, metabolically labeled nascent DNA is site-specifically detected using azido-functionalized PMIP (PMIP-N3) via click chemistry, thereby enabling the super-resolution imaging of nascent DNA in phosphate-buffered saline with a 9-fold improvement in spatial resolution. These results indicate the potential of PMIP-based NIR blinking fluorophores for biological applications of SMLM.


Assuntos
Corantes Fluorescentes , Imagem Individual de Molécula , Animais , Corantes Fluorescentes/química , Microscopia de Fluorescência , Imagem Individual de Molécula/métodos , Imagem Óptica , DNA , Mamíferos
4.
EMBO J ; 37(9)2018 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-29581097

RESUMO

Polymerase-blocking DNA lesions are thought to elicit a checkpoint response via accumulation of single-stranded DNA at stalled replication forks. However, as an alternative to persistent fork stalling, re-priming downstream of lesions can give rise to daughter-strand gaps behind replication forks. We show here that the processing of such structures by an exonuclease, Exo1, is required for timely checkpoint activation, which in turn prevents further gap erosion in S phase. This Rad9-dependent mechanism of damage signaling is distinct from the Mrc1-dependent, fork-associated response to replication stress induced by conditions such as nucleotide depletion or replisome-inherent problems, but reminiscent of replication-independent checkpoint activation by single-stranded DNA Our results indicate that while replisome stalling triggers a checkpoint response directly at the stalled replication fork, the response to replication stress elicited by polymerase-blocking lesions mainly emanates from Exo1-processed, postreplicative daughter-strand gaps, thus offering a mechanistic explanation for the dichotomy between replisome- versus template-induced checkpoint signaling.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Replicação do DNA/fisiologia , DNA Fúngico/biossíntese , Fase S/fisiologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , DNA Fúngico/genética , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
5.
Nucleic Acids Res ; 46(16): 8347-8356, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30107417

RESUMO

Replication of damaged DNA is challenging because lesions in the replication template frequently interfere with an orderly progression of the replisome. In this situation, complete duplication of the genome is ensured by the action of DNA damage bypass pathways effecting either translesion synthesis by specialized, damage-tolerant DNA polymerases or a recombination-like mechanism called template switching (TS). Here we report that budding yeast Pif1, a helicase known to be involved in the resolution of complex DNA structures as well as the maturation of Okazaki fragments during replication, contributes to DNA damage bypass. We show that Pif1 expands regions of single-stranded DNA, so-called daughter-strand gaps, left behind the replication fork as a consequence of replisome re-priming. This function requires interaction with the replication clamp, proliferating cell nuclear antigen, facilitating its recruitment to damage sites, and complements the activity of an exonuclease, Exo1, in the processing of post-replicative daughter-strand gaps in preparation for TS. Our results thus reveal a novel function of a conserved DNA helicase that is known as a key player in genome maintenance.


Assuntos
Dano ao DNA/genética , DNA Helicases/genética , Reparo do DNA/genética , Exodesoxirribonucleases/genética , Proteínas de Saccharomyces cerevisiae/genética , DNA/genética , Replicação do DNA/genética , DNA de Cadeia Simples , DNA Polimerase Dirigida por DNA/genética , Genoma Fúngico/genética , Conformação de Ácido Nucleico , Saccharomyces cerevisiae/genética
6.
Proc Natl Acad Sci U S A ; 114(11): E2205-E2214, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28246327

RESUMO

DNA lesion bypass is mediated by DNA damage tolerance (DDT) pathways and homologous recombination (HR). The DDT pathways, which involve translesion synthesis and template switching (TS), are activated by the ubiquitylation (ub) of PCNA through components of the RAD6-RAD18 pathway, whereas the HR pathway is independent of RAD18 However, it is unclear how these processes are coordinated within the context of chromatin. Here we show that Bre1, an ubiquitin ligase specific for histone H2B, is recruited to chromatin in a manner coupled to replication of damaged DNA. In the absence of Bre1 or H2Bub, cells exhibit accumulation of unrepaired DNA lesions. Consequently, the damaged forks become unstable and resistant to repair. We provide physical, genetic, and cytological evidence that H2Bub contributes toward both Rad18-dependent TS and replication fork repair by HR. Using an inducible system of DNA damage bypass, we further show that H2Bub is required for the regulation of DDT after genome duplication. We propose that Bre1-H2Bub facilitates fork recovery and gap-filling repair by controlling chromatin dynamics in response to replicative DNA damage.


Assuntos
Dano ao DNA , Replicação do DNA , Histonas/metabolismo , Alquilantes/farmacologia , Cromatina/genética , Cromatina/metabolismo , Dano ao DNA/efeitos dos fármacos , Reparo do DNA , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Recombinação Homóloga , Rad51 Recombinase/metabolismo , Origem de Replicação , Transdução de Sinais , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
7.
Nucleic Acids Res ; 39(9): 3632-42, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21227930

RESUMO

The lesion bypass pathway, which is regulated by monoubiquitination of proliferating cell nuclear antigen (PCNA), is essential for resolving replication stalling due to DNA lesions. This process is important for preventing genomic instability and cancer development. Previously, it was shown that cells deficient in tumour suppressor p33ING1 (ING1b) are hypersensitive to DNA damaging agents via unknown mechanism. In this study, we demonstrated a novel tumour suppressive function of ING1b in preserving genomic stability upon replication stress through regulating PCNA monoubiquitination. We found that ING1b knockdown cells are more sensitive to UV due to defects in recovering from UV-induced replication blockage, leading to enhanced genomic instability. We revealed that ING1b is required for the E3 ligase Rad18-mediated PCNA monoubiquitination in lesion bypass. Interestingly, ING1b-mediated PCNA monoubiquitination is associated with the regulation of histone H4 acetylation. Results indicate that chromatin remodelling contributes to the stabilization of stalled replication fork and to the regulation of PCNA monoubiquitination during lesion bypass.


Assuntos
Dano ao DNA , Instabilidade Genômica , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Acetilação , Linhagem Celular , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Técnicas de Silenciamento de Genes , Histonas/metabolismo , Humanos , Proteína 1 Inibidora do Crescimento , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Nucleares/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Fase S , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases , Ubiquitinação , Raios Ultravioleta
8.
Sci Adv ; 9(36): eadf3041, 2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37672592

RESUMO

In eukaryotes, the posttranslational modifier ubiquitin is used to regulate the amounts, interactions, or activities of proteins in diverse pathways and signaling networks. Its effects are mediated by monoubiquitin or polyubiquitin chains of varying geometries. We describe the design, validation, and application of a series of avidity-based probes against the ubiquitylated forms of the DNA replication clamp, proliferating cell nuclear antigen (PCNA), in budding yeast. Directed against total ubiquitylated PCNA or specifically K63-polyubiquitylated PCNA, the probes are tunable in their activities and can be used either as biosensors or as inhibitors of the PCNA-dependent DNA damage bypass pathway. Used in live cells, the probes revealed the timing of PCNA ubiquitylation during damage bypass and a particular susceptibility of the ribosomal DNA locus to the activation of the pathway. Our approach is applicable to a wide range of ubiquitin-conjugated proteins, thus representing a generalizable strategy for the design of biosensors for specific (poly)ubiquitylated forms of individual substrates.


Assuntos
Dano ao DNA , Replicação do DNA , Antígeno Nuclear de Célula em Proliferação , DNA Ribossômico , Ubiquitina
9.
Cell Mol Life Sci ; 68(1): 45-54, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20803232

RESUMO

The Inhibitor of Growth (ING) proteins belong to a well-conserved family which presents in diverse organisms with several structural and functional domains for each protein. The ING family members are found in association with many cellular processes. Thus, the ING family proteins are involved in regulation of gene transcription, DNA repair, tumorigenesis, apoptosis, cellular senescence and cell cycle arrest. The ING proteins have multiple domains that are potentially capable of binding to many partners. It is conceivable, therefore, that such proteins could function similarly within protein complexes. In this case, within this family, each function could be attributed to a specific domain. However, the role of ING domains is not definitively clear. In this review, we summarize recent advances in structure-function relationships in ING proteins. For each domain, we describe the known biological functions and the approaches utilized to identify the functions associated with ING proteins.


Assuntos
Proteínas Supressoras de Tumor/fisiologia , Motivos de Aminoácidos , Montagem e Desmontagem da Cromatina , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/fisiologia , Família Multigênica , Sinais de Localização Nuclear , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Transcrição Gênica , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo
10.
DNA Repair (Amst) ; 105: 103163, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34186497

RESUMO

Dealing with DNA lesions during genome replication is particularly challenging because damaged replication templates interfere with the progression of the replicative DNA polymerases and thereby endanger the stability of the replisome. A variety of mechanisms for the recovery of replication forks exist, but both bacteria and eukaryotic cells also have the option of continuing replication downstream of the lesion, leaving behind a daughter-strand gap in the newly synthesized DNA. In this review, we address the significance of these single-stranded DNA structures as sites of DNA damage sensing and processing at a distance from ongoing genome replication. We describe the factors controlling the emergence of daughter-strand gaps from stalled replication intermediates, the benefits and risks of their expansion and repair via translesion synthesis or recombination-mediated template switching, and the mechanisms by which they activate local as well as global replication stress signals. Our growing understanding of daughter-strand gaps not only identifies them as targets of fundamental genome maintenance mechanisms, but also suggests that proper control over their activities has important practical implications for treatment strategies and resistance mechanisms in cancer therapy.


Assuntos
Dano ao DNA , Reparo do DNA , Replicação do DNA , Transdução de Sinais , Animais , DNA/metabolismo , Eucariotos/genética , Eucariotos/metabolismo , Humanos
11.
Cell Rep ; 36(4): 109440, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34320356

RESUMO

The minichromosome maintenance (MCM) helicase physically interacts with the recombination proteins Rad51 and Rad52 from yeast to human cells. We show, in Saccharomyces cerevisiae, that these interactions occur within a nuclease-insoluble scaffold enriched in replication/repair factors. Rad51 accumulates in a MCM- and DNA-binding-independent manner and interacts with MCM helicases located outside of the replication origins and forks. MCM, Rad51, and Rad52 accumulate in this scaffold in G1 and are released during the S phase. In the presence of replication-blocking lesions, Cdc7 prevents their release from the scaffold, thus maintaining the interactions. We identify a rad51 mutant that is impaired in its ability to bind to MCM but not to the scaffold. This mutant is proficient in recombination but partially defective in single-stranded DNA (ssDNA) gap filling and replication fork progression through damaged DNA. Therefore, cells accumulate MCM/Rad51/Rad52 complexes at specific nuclear scaffolds in G1 to assist stressed forks through non-recombinogenic functions.


Assuntos
Replicação do DNA , DNA de Cadeia Simples/metabolismo , Recombinação Homóloga/genética , Complexos Multiproteicos/metabolismo , Rad51 Recombinase/metabolismo , Saccharomyces cerevisiae/metabolismo , Ciclo Celular/genética , Núcleo Celular/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , Metanossulfonato de Metila , Modelos Biológicos , Ligação Proteica , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Solubilidade
12.
EMBO Rep ; 9(6): 576-81, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18388957

RESUMO

The tumour suppressor p33(ING1b) ((ING1b) for inhibitor of growth family, member 1b) is important in cellular stress responses, including cell-cycle arrest, apoptosis, chromatin remodelling and DNA repair; however, its degradation pathway is still unknown. Recently, we showed that genotoxic stress induces p33(ING1b) phosphorylation at Ser 126, and abolishment of Ser 126 phosphorylation markedly shortened its half-life. Therefore, we suggest that Ser 126 phosphorylation modulates the interaction of p33(ING1b) with its degradation machinery, stabilizing this protein. Combining the use of inhibitors of the main degradation pathways in the nucleus (proteasome and calpains), partial isolation of the proteasome complex, and in vitro interaction and degradation assays, we set out to determine the degradation mechanism of p33(ING1b). We found that p33(ING1b) is degraded in the 20S proteasome and that NAD(P)H quinone oxidoreductase 1 (NQO1), an oxidoreductase previously shown to modulate the degradation of p53 in the 20S proteasome, inhibits the degradation of p33(ING1b). Furthermore, ultraviolet irradiation induces p33(ING1b) phosphorylation at Ser 126, which, in turn, facilitates its interaction with NQO1.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , NAD(P)H Desidrogenase (Quinona)/metabolismo , Proteínas Nucleares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/metabolismo , Animais , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Cicloeximida/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Humanos , Proteína 1 Inibidora do Crescimento , Peptídeos e Proteínas de Sinalização Intracelular/genética , Leupeptinas/metabolismo , NAD(P)H Desidrogenase (Quinona)/genética , Proteínas Nucleares/genética , Fosforilação , Inibidores de Proteassoma , Serina/metabolismo , Proteínas Supressoras de Tumor/genética , Raios Ultravioleta
13.
Oncol Rep ; 19(4): 933-7, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18357378

RESUMO

Myeloid leukemia-1 (Mcl-1) is an anti-apoptotic protein implicated in tumor progression. Its expression was found to be elevated in many types of human cancers and is correlated with tumor progression. The expression of Mcl-1 in melanoma is not fully understood. We investigated the expression of Mcl-1 in normal nevi, dysplastic nevi, primary melanoma and melanoma metastases by tissue microarray and immunohistochemistry. We found that Mcl-1 expression was significantly increased in dysplastic nevi, primary melanoma and melanoma metastases when compared to normal nevi, though the expression of Mcl-1 was decreased in metastatic melanoma when compared to dysplastic nevi. We did not find any correlation between Mcl-1 expression and melanoma patient survival. Our data suggest that Mcl-1 may play a critical role in the initiation of melanoma development.


Assuntos
Síndrome do Nevo Displásico/metabolismo , Melanoma/química , Proteínas de Neoplasias/análise , Nevo/química , Proteínas Proto-Oncogênicas c-bcl-2/análise , Adulto , Idoso , Apoptose , Síndrome do Nevo Displásico/mortalidade , Síndrome do Nevo Displásico/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Melanoma/mortalidade , Melanoma/patologia , Melanoma/secundário , Pessoa de Meia-Idade , Proteína de Sequência 1 de Leucemia de Células Mieloides , Nevo/mortalidade , Nevo/patologia , Análise Serial de Tecidos
14.
Mol Cancer Ther ; 6(6): 1692-700, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17575101

RESUMO

Melanoma is a life-threatening disease with a high mortality rate due to rapid metastasis. Currently, there is no effective treatment for metastatic melanoma. Integrin-linked kinase (ILK) is a serine/threonine kinase and has its role implicated in connecting cell-extracellular matrix interaction and growth factor signaling to cell survival, cell migration, invasion, anchorage-independent growth, angiogenesis, and epithelial-mesenchymal transition. However, the functional role of ILK in melanoma progression is not completely understood. We have previously shown that strong ILK expression was significantly associated with melanoma thickness. In this study, we further elucidate the role of ILK in melanoma cell migration, invasion, anchorage-independent growth, and tumor growth in vivo by specific ILK knockdown using small interfering RNA and short hairpin RNA. We found that ILK knockdown impeded melanoma cell migration, which was associated with reduced stress fiber formation, cell spreading, and cell adhesion. Furthermore, ILK knockdown decreased the invasion ability of melanoma cells and the formation of anchorage-independent colonies in soft agar. Moreover, ILK knockdown significantly impaired the growth of melanoma xenografts in severe combined immunodeficient mice. This study highlights the importance of ILK in melanoma progression and provides an attractive target for the treatment of melanoma.


Assuntos
Divisão Celular/fisiologia , Movimento Celular/fisiologia , Melanoma/patologia , Invasividade Neoplásica , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Sequência de Bases , Linhagem Celular Tumoral , Humanos , Masculino , Melanoma/enzimologia , Camundongos , Camundongos SCID , Oligonucleotídeos , RNA Interferente Pequeno
15.
Front Genet ; 7: 87, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27242895

RESUMO

Complete and faithful duplication of its entire genetic material is one of the essential prerequisites for a proliferating cell to maintain genome stability. Yet, during replication DNA is particularly vulnerable to insults. On the one hand, lesions in replicating DNA frequently cause a stalling of the replication machinery, as most DNA polymerases cannot cope with defective templates. This situation is aggravated by the fact that strand separation in preparation for DNA synthesis prevents common repair mechanisms relying on strand complementarity, such as base and nucleotide excision repair, from working properly. On the other hand, the replication process itself subjects the DNA to a series of hazardous transformations, ranging from the exposure of single-stranded DNA to topological contortions and the generation of nicks and fragments, which all bear the risk of inducing genomic instability. Dealing with these problems requires rapid and flexible responses, for which posttranslational protein modifications that act independently of protein synthesis are particularly well suited. Hence, it is not surprising that members of the ubiquitin family, particularly ubiquitin itself and SUMO, feature prominently in controlling many of the defensive and restorative measures involved in the protection of DNA during replication. In this review we will discuss the contributions of ubiquitin and SUMO to genome maintenance specifically as they relate to DNA replication. We will consider cases where the modifiers act during regular, i.e., unperturbed stages of replication, such as initiation, fork progression, and termination, but also give an account of their functions in dealing with lesions, replication stalling and fork collapse.

16.
Pigment Cell Melanoma Res ; 25(2): 213-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22145991

RESUMO

The E3 ligase Rad18 is a key regulator for the lesion bypass pathway, which plays an important role in genomic stability. However, the status of Rad18 expression in melanoma is not known. Using melanoma tissue microarray (TMA), we showed that nuclear Rad18 expression was upregulated in primary and metastatic melanoma compared to dysplastic nevi. Rad18 expression was significantly reduced in sun-exposed sites compared to the sun-protected sites. Strong Rad18 expression correlated with worse 5-year patient survival and was an independent prognostic factor for melanoma found in the sun-protected sites. Furthermore, we showed that melanoma cell proliferation and the expression of pAkt and cyclin D1 were reduced upon Rad18 knockdown. We, for the first time, showed that Rad18 is significantly increased in melanoma and predicts the poor outcome for melanoma in the sun-protected sites. Rad18 is involved in the regulation of melanoma cell proliferation, which can be exploited in designing new strategy for melanoma treatment.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1 , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Modelos de Riscos Proporcionais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Análise de Sobrevida , Ubiquitina-Proteína Ligases
17.
Curr Drug Targets ; 10(5): 455-64, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19442117

RESUMO

The INhibitor of Growth (ING) genes were discovered during the past decade and identified as type II tumor suppressor genes. Previous studies demonstrated that ING family members participate in various cellular stress responses and thus play important roles in the pathogenesis of various types of cancers, including melanoma. Epidemiological studies showed that UV radiation is the primary etiological factor in melanoma development. Here we review the studies on the role of ING proteins in cellular responses to UV irradiation, melanoma cell motility, and melanoma progression.


Assuntos
Melanoma/fisiopatologia , Proteínas Supressoras de Tumor/metabolismo , Raios Ultravioleta/efeitos adversos , Movimento Celular/fisiologia , Progressão da Doença , Humanos , Melanoma/etiologia , Estresse Fisiológico/fisiologia , Proteínas Supressoras de Tumor/genética
18.
Clin Cancer Res ; 15(20): 6404-11, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19808872

RESUMO

PURPOSE: Aberrant expression of SWI/SNF chromatin remodeling complex is involved in cancer development. The tumor suppressor SNF5, the core subunit of SWI/SNF complex, has been shown to regulate cell differentiation, cell cycle control, and apoptosis. To investigate the role of SNF5 in the development of melanoma, we examined the expression of SNF5 in melanocytic lesions at different stages and analyzed the correlation between SNF5 expression and clinicopathologic variables and patient survival. EXPERIMENTAL DESIGN: Using tissue microarray and immunohistochemistry, we evaluated SNF5 staining in 51 dysplastic nevi, 88 primary melanomas, and 48 metastatic melanomas. We studied chemosensitivity of melanoma cells with reduced SNF5 expression by siRNA using cell survival and apoptosis assays. RESULTS: SNF5 expression was reduced in metastatic melanoma compared with dysplastic nevi (P = 0.005), in advanced primary melanoma (Clark's level V) compared with low risk Clark's level II melanoma (P = 0.019), and in melanoma at sun-exposed sites compared with sun-protected sites (P = 0.044). Furthermore, we showed a strong correlation between negative SNF5 expression and a worse 5-year survival in melanoma patients (P = 0.016). Multivariate Cox regression analysis revealed that negative SNF5 expression is an independent prognostic factor to predict patient outcome in primary melanomas (P = 0.031). Finally, we showed that knockdown of SNF5 in melanoma cell lines resulted in significant chemoresistance. CONCLUSIONS: Our data indicate that SNF5 may be an important marker for human melanoma progression and prognosis as well as a potential therapeutic target.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neoplasias Cutâneas/metabolismo , Fatores de Transcrição/metabolismo , Biomarcadores Tumorais/análise , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Melanoma/genética , Pessoa de Meia-Idade , Prognóstico , Proteína SMARCB1 , Neoplasias Cutâneas/mortalidade , Análise Serial de Tecidos
19.
Exp Cell Res ; 313(8): 1628-38, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17379210

RESUMO

ING1b is the most studied ING family protein and perhaps the most ubiquitously and abundantly expressed. This protein is involved in the regulation of various biological functions ranging from senescence, cell cycle arrest, apoptosis, to DNA repair. ING1b is upregulated by UV irradiation and enhances the removal of bulky nucleic acid photoproducts. In this study, we provide evidence that ING1b mediates nucleotide excision repair by facilitating the access to damaged nucleosomal DNA. We demonstrate that ING1b is not recruited to UV-induced DNA lesions but enhances nucleotide excision repair only in XPC-proficient cells, implying an essential role in early steps of the 'access, repair, restore' model. We also find that ING1b alters histone acetylation dynamics upon exposure to UV radiation and induces chromatin relaxation in microccocal nuclease digestion assay, revealing that ING1b may allow better access to nucleotide excision repair machinery. More importantly, ING1b associates with chromatin in a UV-inducible manner and facilitates DNA access to nucleotide excision repair factor XPA. Furthermore, depletion of the endogenous ING1b results to the sensitization of cells at S-phase to UV irradiation. Taken together, these observations establish a role of ING1b acting as a chromatin accessibility factor for DNA damage recognition proteins upon genotoxic injury.


Assuntos
Cromatina/metabolismo , Reparo do DNA , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteína de Xeroderma Pigmentoso Grupo A/metabolismo , Acetilação , Linhagem Celular , Cromatina/genética , Dano ao DNA , Histonas/metabolismo , Humanos , Proteína 1 Inibidora do Crescimento , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Nucleares/genética , Nucleossomos/genética , Nucleossomos/metabolismo , Proteínas Supressoras de Tumor/genética , Raios Ultravioleta , Proteína de Xeroderma Pigmentoso Grupo A/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa