Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Commun ; 12(1): 512, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33479235

RESUMO

To achieve replicative immortality, cancer cells must activate telomere maintenance mechanisms to prevent telomere shortening. ~85% of cancers circumvent telomeric attrition by re-expressing telomerase, while the remaining ~15% of cancers induce alternative lengthening of telomeres (ALT), which relies on break-induced replication (BIR) and telomere recombination. Although ALT tumours were first reported over 20 years ago, the mechanism of ALT induction remains unclear and no study to date has described a cell-based model that permits the induction of ALT. Here, we demonstrate that infection with Kaposi's sarcoma herpesvirus (KSHV) induces sustained acquisition of ALT-like features in previously non-ALT cell lines. KSHV-infected cells acquire hallmarks of ALT activity that are also observed in KSHV-associated tumour biopsies. Down-regulating BIR impairs KSHV latency, suggesting that KSHV co-opts ALT for viral functionality. This study uncovers KSHV infection as a means to study telomere maintenance by ALT and reveals features of ALT in KSHV-associated tumours.


Assuntos
Neoplasias/genética , Homeostase do Telômero/genética , Encurtamento do Telômero/genética , Telômero/genética , Carcinogênese , Linhagem Celular , Linhagem Celular Tumoral , Dano ao DNA , Replicação do DNA/genética , Células HeLa , Herpesvirus Humano 8/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Hibridização in Situ Fluorescente , Neoplasias/patologia , Neoplasias/virologia , Proteoma/genética , Proteoma/metabolismo , Telomerase/genética , Telomerase/metabolismo
2.
Nature ; 575(7783): 523-527, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31723267

RESUMO

The protection of telomere ends by the shelterin complex prevents DNA damage signalling and promiscuous repair at chromosome ends. Evidence suggests that the 3' single-stranded telomere end can assemble into a lasso-like t-loop configuration1,2, which has been proposed to safeguard chromosome ends from being recognized as DNA double-strand breaks2. Mechanisms must also exist to transiently disassemble t-loops to allow accurate telomere replication and to permit telomerase access to the 3' end to solve the end-replication problem. However, the regulation and physiological importance of t-loops in the protection of telomere ends remains unknown. Here we identify a CDK phosphorylation site in the shelterin subunit at Ser365 of TRF2, whose dephosphorylation in S phase by the PP6R3 phosphatase provides a narrow window during which the RTEL1 helicase can transiently access and unwind t-loops to facilitate telomere replication. Re-phosphorylation of TRF2 at Ser365 outside of S phase is required to release RTEL1 from telomeres, which not only protects t-loops from promiscuous unwinding and inappropriate activation of ATM, but also counteracts replication conflicts at DNA secondary structures that arise within telomeres and across the genome. Hence, a phospho-switch in TRF2 coordinates the assembly and disassembly of t-loops during the cell cycle, which protects telomeres from replication stress and an unscheduled DNA damage response.


Assuntos
Ciclo Celular , Quinases Ciclina-Dependentes/metabolismo , Fosfosserina/metabolismo , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/química , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , DNA/biossíntese , DNA/química , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Dano ao DNA , DNA Helicases/metabolismo , Reparo do DNA , Replicação do DNA , Fibroblastos , Genoma/genética , Células HEK293 , Humanos , Camundongos , Mutação , Fenótipo , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Antígeno Nuclear de Célula em Proliferação/metabolismo , Fase S , Complexo Shelterina , Telomerase/metabolismo , Telômero/genética , Proteínas de Ligação a Telômeros/química , Proteínas de Ligação a Telômeros/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética
3.
Mol Cell ; 69(2): 292-305.e6, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29351848

RESUMO

Erroneous DNA repair by heterologous recombination (Ht-REC) is a potential threat to genome stability, but evidence supporting its prevalence is lacking. Here we demonstrate that recombination is possible between heterologous sequences and that it is a source of chromosomal alterations in mitotic and meiotic cells. Mechanistically, we find that the RTEL1 and HIM-6/BLM helicases and the BRCA1 homolog BRC-1 counteract Ht-REC in Caenorhabditis elegans, whereas mismatch repair does not. Instead, MSH-2/6 drives Ht-REC events in rtel-1 and brc-1 mutants and excessive crossovers in rtel-1 mutant meioses. Loss of vertebrate Rtel1 also causes a variety of unusually large and complex structural variations, including chromothripsis, breakage-fusion-bridge events, and tandem duplications with distant intra-chromosomal insertions, whose structure are consistent with a role for RTEL1 in preventing Ht-REC during break-induced replication. Our data establish Ht-REC as an unappreciated source of genome instability that underpins a novel class of complex genome rearrangements that likely arise during replication stress.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , DNA Helicases/metabolismo , Instabilidade Genômica/fisiologia , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , DNA Helicases/genética , Reparo de Erro de Pareamento de DNA , Reparo do DNA/genética , Replicação do DNA , Instabilidade Genômica/genética , Mutação , RecQ Helicases/metabolismo , Recombinação Genética/genética
5.
EMBO Rep ; 18(3): 437-450, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28174209

RESUMO

Protein phosphatase 2A (PP2A) is a critical human tumor suppressor. Cancerous inhibitor of PP2A (CIP2A) supports the activity of several critical cancer drivers (Akt, MYC, E2F1) and promotes malignancy in most cancer types via PP2A inhibition. However, the 3D structure of CIP2A has not been solved, and it remains enigmatic how it interacts with PP2A. Here, we show by yeast two-hybrid assays, and subsequent validation experiments, that CIP2A forms homodimers. The homodimerization of CIP2A is confirmed by solving the crystal structure of an N-terminal CIP2A fragment (amino acids 1-560) at 3.0 Å resolution, and by subsequent structure-based mutational analyses of the dimerization interface. We further describe that the CIP2A dimer interacts with the PP2A subunits B56α and B56γ. CIP2A binds to the B56 proteins via a conserved N-terminal region, and dimerization promotes B56 binding. Intriguingly, inhibition of either CIP2A dimerization or B56α/γ expression destabilizes CIP2A, indicating opportunities for controlled degradation. These results provide the first structure-function analysis of the interaction of CIP2A with PP2A/B56 and have direct implications for its targeting in cancer therapy.


Assuntos
Autoantígenos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Oncogênicas/metabolismo , Proteína Fosfatase 2/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Autoantígenos/química , Sítios de Ligação , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/química , Modelos Moleculares , Mutação , Proteínas Oncogênicas/química , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Multimerização Proteica , Proteína Fosfatase 2/química , Proteína Fosfatase 2/genética , Estabilidade Proteica , Subunidades Proteicas/metabolismo , Relação Estrutura-Atividade , Proteínas Supressoras de Tumor/química
6.
Mol Cell ; 64(4): 688-703, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27871365

RESUMO

Covalent DNA-protein crosslinks (DPCs) are toxic DNA lesions that interfere with essential chromatin transactions, such as replication and transcription. Little was known about DPC-specific repair mechanisms until the recent identification of a DPC-processing protease in yeast. The existence of a DPC protease in higher eukaryotes is inferred from data in Xenopus laevis egg extracts, but its identity remains elusive. Here we identify the metalloprotease SPRTN as the DPC protease acting in metazoans. Loss of SPRTN results in failure to repair DPCs and hypersensitivity to DPC-inducing agents. SPRTN accomplishes DPC processing through a unique DNA-induced protease activity, which is controlled by several sophisticated regulatory mechanisms. Cellular, biochemical, and structural studies define a DNA switch triggering its protease activity, a ubiquitin switch controlling SPRTN chromatin accessibility, and regulatory autocatalytic cleavage. Our data also provide a molecular explanation on how SPRTN deficiency causes the premature aging and cancer predisposition disorder Ruijs-Aalfs syndrome.


Assuntos
Proteínas de Caenorhabditis elegans/química , Reparo do DNA , Proteínas de Ligação a DNA/química , DNA/química , Proteínas de Schizosaccharomyces pombe/química , Proteína de Xeroderma Pigmentoso Grupo A/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/efeitos da radiação , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Linhagem Celular , Cisplatino/química , Reagentes de Ligações Cruzadas/química , Cristalografia por Raios X , DNA/genética , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/efeitos da radiação , Formaldeído/química , Células HeLa , Humanos , Cinética , Camundongos , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Schizosaccharomyces/enzimologia , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Raios Ultravioleta , Proteína de Xeroderma Pigmentoso Grupo A/genética , Proteína de Xeroderma Pigmentoso Grupo A/metabolismo
7.
PLoS Pathog ; 12(2): e1005424, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26891221

RESUMO

Kaposi's sarcoma herpesvirus (KSHV) causes Kaposi's sarcoma and certain lymphoproliferative malignancies. Latent infection is established in the majority of tumor cells, whereas lytic replication is reactivated in a small fraction of cells, which is important for both virus spread and disease progression. A siRNA screen for novel regulators of KSHV reactivation identified the E3 ubiquitin ligase MDM2 as a negative regulator of viral reactivation. Depletion of MDM2, a repressor of p53, favored efficient activation of the viral lytic transcription program and viral reactivation. During lytic replication cells activated a p53 response, accumulated DNA damage and arrested at G2-phase. Depletion of p21, a p53 target gene, restored cell cycle progression and thereby impaired the virus reactivation cascade delaying the onset of virus replication induced cytopathic effect. Herpesviruses are known to reactivate in response to different kinds of stress, and our study now highlights the molecular events in the stressed host cell that KSHV has evolved to utilize to ensure efficient viral lytic replication.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , Regulação Viral da Expressão Gênica/genética , Herpesvirus Humano 8/genética , Estresse Fisiológico/genética , Replicação Viral , Linhagem Celular Tumoral , Replicação do DNA , Humanos , RNA Interferente Pequeno/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Ativação Viral/fisiologia , Latência Viral/genética , Replicação Viral/genética
9.
Nat Struct Mol Biol ; 22(11): 867-74, 2015 11.
Artigo em Inglês | MEDLINE | ID: mdl-26581521

RESUMO

Mutations in genes encoding proteins required for telomere structure, replication, repair and length maintenance are associated with several debilitating human genetic disorders. These complex telomere biology disorders (TBDs) give rise to critically short telomeres that affect the homeostasis of multiple organs. Furthermore, genome instability is often a hallmark of telomere syndromes, which are associated with increased cancer risk. Here, we summarize the molecular causes and cellular consequences of disease-causing mutations associated with telomere dysfunction.


Assuntos
Doenças Genéticas Inatas/genética , Homeostase do Telômero , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo , Telômero/metabolismo , Genética Médica , Instabilidade Genômica , Humanos , Mutação
10.
Mol Cell ; 57(4): 622-635, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25620558

RESUMO

The helicase RTEL1 promotes t-loop unwinding and suppresses telomere fragility to maintain the integrity of vertebrate telomeres. An interaction between RTEL1 and PCNA is important to prevent telomere fragility, but how RTEL1 engages with the telomere to promote t-loop unwinding is unclear. Here, we establish that the shelterin protein TRF2 recruits RTEL1 to telomeres in S phase, which is required to prevent catastrophic t-loop processing by structure-specific nucleases. We show that the TRF2-RTEL1 interaction is mediated by a metal-coordinating C4C4 motif in RTEL1, which is compromised by the Hoyeraal-Hreidarsson syndrome (HHS) mutation, RTEL1(R1264H). Conversely, we define a TRF2(I124D) substitution mutation within the TRFH domain of TRF2, which eliminates RTEL1 binding and phenocopies the RTEL1(R1264H) mutation, giving rise to aberrant t-loop excision, telomere length heterogeneity, and loss of the telomere as a circle. These results implicate TRF2 in the recruitment of RTEL1 to facilitate t-loop disassembly at telomeres in S phase.


Assuntos
DNA Helicases/fisiologia , Modelos Genéticos , Fase S , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/fisiologia , Animais , Células Cultivadas , DNA Helicases/química , DNA Helicases/metabolismo , Humanos , Metáfase , Camundongos , Estrutura Terciária de Proteína , Transporte Proteico , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
11.
Trends Cell Biol ; 24(7): 416-25, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24582487

RESUMO

DNA secondary structures that arise during DNA replication, repair, and recombination (3R) must be processed correctly to prevent genetic instability. Regulator of telomere length 1 (RTEL1) is an essential DNA helicase that disassembles a variety of DNA secondary structures to facilitate 3R processes and to maintain telomere integrity. The past few years have witnessed the emergence of RTEL1 variants that confer increased susceptibility to high-grade glioma, astrocytomas, and glioblastomas. Mutations in RTEL1 have also been implicated in Hoyeraal-Hreidarsson syndrome, a severe form of the bone-marrow failure and cancer predisposition disorder, dyskeratosis congenita. We review these recent findings and highlight its crucial link between DNA secondary-structure metabolism and human disease.


Assuntos
DNA Helicases/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Telômero/metabolismo , Animais , DNA Helicases/genética , Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/metabolismo , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Microcefalia/genética , Microcefalia/metabolismo , Mutação/genética , Telômero/genética
12.
PLoS Genet ; 9(8): e1003695, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24009516

RESUMO

Dyskeratosis congenita (DC) is a heterogeneous inherited bone marrow failure and cancer predisposition syndrome in which germline mutations in telomere biology genes account for approximately one-half of known families. Hoyeraal Hreidarsson syndrome (HH) is a clinically severe variant of DC in which patients also have cerebellar hypoplasia and may present with severe immunodeficiency and enteropathy. We discovered a germline autosomal recessive mutation in RTEL1, a helicase with critical telomeric functions, in two unrelated families of Ashkenazi Jewish (AJ) ancestry. The affected individuals in these families are homozygous for the same mutation, R1264H, which affects three isoforms of RTEL1. Each parent was a heterozygous carrier of one mutant allele. Patient-derived cell lines revealed evidence of telomere dysfunction, including significantly decreased telomere length, telomere length heterogeneity, and the presence of extra-chromosomal circular telomeric DNA. In addition, RTEL1 mutant cells exhibited enhanced sensitivity to the interstrand cross-linking agent mitomycin C. The molecular data and the patterns of inheritance are consistent with a hypomorphic mutation in RTEL1 as the underlying basis of the clinical and cellular phenotypes. This study further implicates RTEL1 in the etiology of DC/HH and immunodeficiency, and identifies the first known homozygous autosomal recessive disease-associated mutation in RTEL1.


Assuntos
DNA Helicases/genética , Disceratose Congênita/genética , Disceratose Congênita/patologia , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/patologia , Síndromes de Imunodeficiência/patologia , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Microcefalia/genética , Microcefalia/patologia , Adulto , Disceratose Congênita/etiologia , Feminino , Retardo do Crescimento Fetal/etiologia , Genes Recessivos , Mutação em Linhagem Germinativa , Homozigoto , Humanos , Síndromes de Imunodeficiência/genética , Deficiência Intelectual/etiologia , Judeus , Microcefalia/etiologia , Dados de Sequência Molecular , Mutação , Fenótipo , Telomerase/genética , Telômero/genética
13.
J Virol ; 86(11): 5974-91, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22438557

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is etiologically associated with the angioproliferative Kaposi's sarcoma (KS). KSHV infection and the expression of latency-associated nuclear antigen (LANA-1) upregulates the angiogenic multifunctional 123-amino-acid, 14-kDa protein angiogenin (ANG), which is detected in KS lesions and in KSHV-associated primary effusion lymphoma (PEL) cells. ANG knockdown or the inhibition of ANG's nuclear translocation resulted in decreased LANA-1 gene expression and reduced KSHV-infected endothelial and PEL cell survival (Sadagopan et al., J. Virol. 83:3342-3364, 2009). Further studies here demonstrate that LANA-1 and ANG colocalize and coimmunoprecipitate in de novo infected endothelial cells and in latently infected PEL (BCBL-1 and BC-3) cells. LANA-1 and ANG interaction occurred in the absence of the KSHV genome and other viral proteins. In gel filtration chromatography analyses of BC-3 cell lysates, ANG coeluted with LANA-1, p53, and Mdm2 in high-molecular-weight fractions, and LANA-1, p53, and Mdm2 also coimmunoprecipitated with ANG. LANA-1, ANG, and p53 colocalized in KSHV-infected cells, and colocalization between ANG and p53 was also observed in LANA-1-negative cells. The deletion constructs of ANG suggested that the C-terminal region of amino acids 104 to 123 is involved in LANA-1 and p53 interactions. Silencing ANG or inhibiting its nuclear translocation resulted in decreased nuclear LANA-1 and ANG levels, decreased interactions between ANG-LANA-1, ANG-p53, and LANA-1-p53, the induction of p53, p21, and Bax proteins, the increased cytoplasmic localization of p53, the downregulation of Bcl-2, the increased cleavage of caspase-3, and the apoptosis of cells. No such effects were observed in KSHV-negative BJAB cells. The phosphorylation of p53 at serine 15, which is essential for p53 stabilization and for p53's apoptotic and cell cycle regulation functions, was increased in BCBL-1 cells transduced with short hairpin RNA targeting ANG. Together, these studies suggest that the antiapoptosis observed in KSHV-infected cells and the suppression of p53 functions are mediated in part by ANG, and KSHV has probably evolved to utilize angiogenin's multiple functions for the maintenance of its latency and cell survival. Thus, targeting ANG to induce the apoptosis of cells latently infected with KSHV is an attractive therapeutic strategy against KSHV infection and associated malignancies.


Assuntos
Antígenos Virais/metabolismo , Apoptose , Herpesvirus Humano 8/patogenicidade , Interações Hospedeiro-Patógeno , Proteínas Nucleares/metabolismo , Ribonuclease Pancreático/metabolismo , Linhagem Celular , Cromatografia em Gel , Células Endoteliais/virologia , Humanos , Imunoprecipitação , Mapeamento de Interação de Proteínas , Deleção de Sequência
14.
Cell Host Microbe ; 10(6): 577-90, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22177562

RESUMO

Kaposi sarcoma (KS), an angioproliferative disease associated with Kaposi sarcoma herpesvirus (KSHV) infection, harbors a diversity of cell types ranging from endothelial to mesenchymal cells of unclear origin. We developed a three-dimensional cell model for KSHV infection and used it to demonstrate that KSHV induces transcriptional reprogramming of lymphatic endothelial cells to mesenchymal cells via endothelial-to-mesenchymal transition (EndMT). KSHV-induced EndMT was initiated by the viral proteins vFLIP and vGPCR through Notch pathway activation, leading to gain of membrane-type-1 matrix metalloproteinase (MT1-MMP)-dependent invasive properties and concomitant changes in viral gene expression. Mesenchymal markers and MT1-MMP were found codistributed with a KSHV marker in the same cells from primary KS biopsies. Our data explain the heterogeneity of cell types within KS lesions and suggest that KSHV-induced EndMT may contribute to KS development by giving rise to infected, invasive cells while providing the virus a permissive cellular microenvironment for efficient spread.


Assuntos
Transição Epitelial-Mesenquimal , Herpesvirus Humano 8/fisiologia , Metaloproteinase 14 da Matriz/metabolismo , Receptores Notch/metabolismo , Sarcoma de Kaposi/enzimologia , Sarcoma de Kaposi/fisiopatologia , Linhagem Celular , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Células Endoteliais/virologia , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/genética , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/enzimologia , Células-Tronco Mesenquimais/virologia , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Transdução de Sinais , Proteínas Virais/genética , Proteínas Virais/metabolismo
15.
PLoS Pathog ; 6(3): e1000818, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20333249

RESUMO

Nucleophosmin (NPM) is a multifunctional nuclear phosphoprotein and a histone chaperone implicated in chromatin organization and transcription control. Oncogenic Kaposi's sarcoma herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma, primary effusion lymphoma (PEL) and multicentric Castleman disease (MCD). In the infected host cell KSHV displays two modes of infection, the latency and productive viral replication phases, involving extensive viral DNA replication and gene expression. A sustained balance between latency and reactivation to the productive infection state is essential for viral persistence and KSHV pathogenesis. Our study demonstrates that the KSHV v-cyclin and cellular CDK6 kinase phosphorylate NPM on threonine 199 (Thr199) in de novo and naturally KSHV-infected cells and that NPM is phosphorylated to the same site in primary KS tumors. Furthermore, v-cyclin-mediated phosphorylation of NPM engages the interaction between NPM and the latency-associated nuclear antigen LANA, a KSHV-encoded repressor of viral lytic replication. Strikingly, depletion of NPM in PEL cells leads to viral reactivation, and production of new infectious virus particles. Moreover, the phosphorylation of NPM negatively correlates with the level of spontaneous viral reactivation in PEL cells. This work demonstrates that NPM is a critical regulator of KSHV latency via functional interactions with v-cyclin and LANA.


Assuntos
Quinase 6 Dependente de Ciclina/metabolismo , Herpesvirus Humano 8/crescimento & desenvolvimento , Proteínas Nucleares/metabolismo , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Latência Viral/fisiologia , Acetilação , Antígenos Virais/genética , Antígenos Virais/metabolismo , Linhagem Celular Tumoral , Herpesvirus Humano 8/genética , Humanos , Proteínas Nucleares/genética , Nucleofosmina , Fosforilação/fisiologia , RNA Interferente Pequeno , Treonina/metabolismo , Replicação Viral/fisiologia
16.
Cell Cycle ; 6(18): 2205-9, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17890905

RESUMO

KSHV infection is the causative agent in three different tumor types: Kaposi's sarcoma, a plasmablastic variant of multicentric Castelman's disease and an AIDS-related form of B cell lymphoproliferative disorder called primary effusion lymphoma (PEL). PEL manifests as an effusion malignancy in Kaposi's sarcoma patients with advanced AIDS, but also occurs in HIV-negative individuals. PEL is a very aggressive disease, and currently there are no efficient therapies for treating PEL. In our recent paper we report that p53 reactivation by a small molecule inhibitor of p53-MDM2 interaction, Nutlin-3a, induces selective and massive apoptosis in PEL cells, and has striking anti-tumor activity in a mouse xenograft PEL model. In the light of current treatment regimens for PEL, we discuss here the benefits of using reactivation of the p53 pathway as a novel principle for the treatment of this virally induced highly aggressive malignancy.


Assuntos
Herpesvirus Humano 8/patogenicidade , Linfoma Relacionado a AIDS/terapia , Linfoma Relacionado a AIDS/virologia , Sarcoma de Kaposi/terapia , Sarcoma de Kaposi/virologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Herpesvirus Humano 8/crescimento & desenvolvimento , Humanos , Linfoma Relacionado a AIDS/metabolismo , Linfoma Relacionado a AIDS/patologia , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patologia , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/fisiologia
17.
J Clin Invest ; 117(4): 1019-28, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17364023

RESUMO

Kaposi's sarcoma herpesvirus (KSHV) is the etiologic agent for primary effusion lymphoma (PEL), a non-Hodgkin type lymphoma manifesting as an effusion malignancy in the affected individual. Although KSHV has been recognized as a tumor virus for over a decade, the pathways for its tumorigenic conversion are incompletely understood, which has greatly hampered the development of efficient therapies for KSHV-induced malignancies like PEL and Kaposi's sarcoma. There are no current therapies effective against the aggressive, KSHV-induced PEL. Here we demonstrate that activation of the p53 pathway using murine double minute 2 (MDM2) inhibitor Nutlin-3a conveyed specific and highly potent activation of PEL cell killing. Our results demonstrated that the KSHV latency-associated nuclear antigen (LANA) bound to both p53 and MDM2 and that the MDM2 inhibitor Nutlin-3a disrupted the p53-MDM2-LANA complex and selectively induced massive apoptosis in PEL cells. Together with our results indicating that KSHV-infection activated DNA damage signaling, these findings contribute to the specificity of the cytotoxic effects of Nutlin-3a in KSHV-infected cells. Moreover, we showed that Nutlin-3a had striking antitumor activity in vivo in a mouse xenograft model. Our results therefore present new options for exploiting reactivation of p53 as what we believe to be a novel and highly selective treatment modality for this virally induced lymphoma.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genes p53 , Herpesvirus Humano 8/fisiologia , Linfoma/virologia , Sarcoma de Kaposi/genética , Proteína Supressora de Tumor p53/genética , Animais , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/genética , Herpesvirus Humano 4/patogenicidade , Herpesvirus Humano 4/fisiologia , Herpesvirus Humano 8/patogenicidade , Humanos , Imidazóis/farmacologia , Linfoma/genética , Camundongos , Piperazinas/farmacologia , Sarcoma de Kaposi/virologia , Transplante Heterólogo , Latência Viral
18.
Mol Cell Biol ; 26(6): 2430-40, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16508017

RESUMO

K cyclin encoded by Kaposi's sarcoma-associated herpesvirus confers resistance to the cyclin-dependent kinase (cdk) inhibitors p16Ink4A, p21Cip1, and p27Kip1 on the associated cdk6. We have previously shown that K cyclin expression enforces S-phase entry on cells overexpressing p27Kip1 by promoting phosphorylation of p27Kip1 on threonine 187, triggering p27Kip1 down-regulation. Since p21Cip1 acts in a manner similar to that of p27Kip1, we have investigated the subversion of a p21Cip1-induced G1 arrest by K cyclin. Here, we show that p21Cip1 is associated with K cyclin both in overexpression models and in primary effusion lymphoma cells and is a substrate of the K cyclin/cdk6 complex, resulting in phosphorylation of p21Cip1 on serine 130. This phosphoform of p21Cip1 appeared unable to associate with cdk2 in vivo. We further demonstrate that phosphorylation on serine 130 is essential for K cyclin-mediated release of a p21Cip1-imposed G1 arrest. Moreover, we show that under physiological conditions of cell cycle arrest due to elevated levels of p21Cip1 resulting from oxidative stress, K cyclin expression enabled S-phase entry and was associated with p21Cip1 phosphorylation and partial restoration of cdk2 kinase activity. Thus, expression of the viral cyclin enables cells to subvert the cell cycle inhibitory function of p21Cip1 by promoting cdk6-dependent phosphorylation of this antiproliferative protein.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Ciclinas/metabolismo , Fase G1/fisiologia , Serina/metabolismo , Proteínas Virais/metabolismo , Células 3T3/metabolismo , Animais , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Ciclinas/genética , Camundongos , Estresse Oxidativo , Fosforilação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Virais/genética
19.
Blood ; 107(2): 725-32, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16160006

RESUMO

Kaposi sarcoma herpesvirus (KSHV) infection is consistently associated with primary effusion lymphomas (PELs) that are non-Hodgkin lymphomas of B-cell origin. All PEL cells are latently infected with KSHV and express latent viral proteins such as the viral cyclin (v-cyclin), which has previously been implicated in down-regulation of cell-cycle inhibitor p27(KIP1) levels via phosphorylation on Thr187. PEL cells retain high levels of p27(KIP1) but yet proliferate actively, which has left the biologic significance of this p27(KIP1) destabilization somewhat elusive. We have recently demonstrated that v-cyclin and p27(KIP1) stably associate in PEL cells. Here we demonstrate that v-cyclin together with its kinase partner CDK6 phosphorylates the associated p27(KIP1) in PEL cells, which represent a biologically relevant model system for KSHV pathobiology. During latent viral replication p27(KIP1) was phosphorylated by v-cyclin-CDK6 predominantly on Ser10, which enhances its cytoplasmic localization. Interestingly, upon reactivation of KSHV lytic cycle, v-cyclin-CDK6 phosphorylated p27(KIP1) on Thr187, which resulted in down-regulation of p27(KIP1) protein levels. These findings indicate that v-cyclin modulates the cell-cycle inhibitory function of p27(KIP1) by phosphorylation in PELs, and also suggest a novel role for v-cyclin in the lytic reactivation of KSHV.


Assuntos
Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p27/antagonistas & inibidores , Herpesvirus Humano 8/metabolismo , Linfoma Relacionado a AIDS/metabolismo , Sarcoma de Kaposi/metabolismo , Proteínas Virais/farmacologia , Quinase 6 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Ciclinas/farmacologia , Citoplasma/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Humanos , Immunoblotting , Imunoprecipitação , Linfoma Relacionado a AIDS/virologia , Fosforilação , Transporte Proteico , Sarcoma de Kaposi/patologia , Sarcoma de Kaposi/virologia , Serina/química , Frações Subcelulares , Treonina/química , Células Tumorais Cultivadas , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA