Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
1.
Oncogene ; 37(25): 3329-3339, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29551768

RESUMO

Chronic liver injury triggers liver fibrosis and hepatocellular carcinoma (HCC), the third leading cause of cancer-related mortality. Cyclin E1 (CcnE1, formerly designated Cyclin E) is a regulatory subunit of the Cyclin-dependent kinase 2 (CDK2). It is overexpressed in approximately 70% of human HCCs correlating with poor prognosis, while the relevance of its orthologue Cyclin E2 (CcnE2) is unclear. Hepatocyte-specific deletion of NF-kappa-B essential modulator (NEMOΔhepa) leads to chronic hepatitis, liver fibrosis, and HCC as well as CcnE upregulation. To this end, we generated NEMOΔhepa/CcnE1-/- and NEMOΔhepa/CcnE2-/- double knockout mice and investigated age-dependent liver disease progression in these animals. Deletion of CcnE1 in NEMOΔhepa mice decreased basal liver damage and reduced spontaneous liver inflammation in young mice. In contrast, loss of CcnE2 did not affect liver injury in NEMOΔhepa livers pointing to a unique, non-redundant function of CcnE1 in chronic hepatitis. Accordingly, basal compensatory hepatocyte proliferation in NEMOΔhepa mice was reduced by concomitant ablation of CcnE1, but not after loss of CcnE2. In aged NEMOΔhepa mice, loss of CcnE1 resulted in significant reduction of liver tumorigenesis, while deletion of CcnE2 had no effect on HCC formation. CcnE1, but not its orthologue CcnE2, substantially contributes to hepatic inflammatory response, liver disease progression, and hepatocarcinogenesis in NEMOΔhepa mice.


Assuntos
Carcinoma Hepatocelular/prevenção & controle , Ciclina E/fisiologia , Ciclinas/fisiologia , Modelos Animais de Doenças , Hepatite/prevenção & controle , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neoplasias Hepáticas Experimentais/prevenção & controle , Fígado/lesões , Fígado/patologia , Proteínas Oncogênicas/fisiologia , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/patologia , Hepatite/etiologia , Hepatite/patologia , Neoplasias Hepáticas Experimentais/etiologia , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
J Biol Chem ; 292(52): 21264-21281, 2017 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-29109143

RESUMO

The tumor microenvironment is characterized by nutrient-deprived conditions in which the cancer cells have to adapt for survival. Serum starvation resembles the growth factor deprivation characteristic of the poorly vascularized tumor microenvironment and has aided in the discovery of key growth regulatory genes and microRNAs (miRNAs) that have a role in the oncogenic transformation. We report here that miR-874 down-regulates the major G1/S phase cyclin, cyclin E1 (CCNE1), during serum starvation. Because the adaptation of cancer cells to the tumor microenvironment is vital for subsequent oncogenesis, we tested for miR-874 and CCNE1 interdependence in osteosarcoma cells. We observed that miR-874 inhibits CCNE1 expression in primary osteoblasts, but in aggressive osteosarcomas, miR-874 is down-regulated, leading to elevated CCNE1 expression and appearance of cancer-associated phenotypes. We established that loss of miR-874-mediated control of cyclin E1 is a general feature of osteosarcomas. The down-regulation of CCNE1 by miR-874 is independent of E2F transcription factors. Restoration of miR-874 expression impeded S phase progression, suppressing aggressive growth phenotypes, such as cell invasion, migration, and xenograft tumors, in nude mice. In summary, we report that miR-874 inhibits CCNE1 expression during growth factor deprivation and that miR-874 down-regulation in osteosarcomas leads to CCNE1 up-regulation and more aggressive growth phenotypes.


Assuntos
Ciclina E/fisiologia , MicroRNAs/fisiologia , Proteínas Oncogênicas/fisiologia , Osteossarcoma/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Ciclina E/genética , Ciclina G1/metabolismo , Regulação para Baixo , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Oncogênicas/genética , Oncogenes , Osteossarcoma/genética , Fase S
3.
Microsc Microanal ; 23(1): 69-76, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28162122

RESUMO

Cyclin E1 (CCNE1) is a core component of cell cycle regulation that drives the transition into the S phase. CCNE1 plays critical roles in cell cycle, cell proliferation, and cellular functions. However, the function of CCNE1 in early embryonic development is limited. In the present study, the function and expression of Ccne1 in porcine early parthenotes were examined. Immunostaining experiments showed that CCNE1 localized in the nucleus, starting at the four-cell stage. Knockdown of Ccne1 by double-stranded RNA resulted in the failure of blastocyst formation and induced blastocyst apoptosis. Ccne1 depletion increased expression of the pro-apoptotic gene Bax, and decreased the expression of Oct4 and the rate of inner cell mass (ICM)/trophectoderm formation. The results indicated that CCNE1 affects blastocyst formation by inducing cell apoptosis and ICM formation during porcine embryonic development.


Assuntos
Ciclina E/farmacologia , Ciclina E/fisiologia , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/fisiologia , Microscopia de Fluorescência/métodos , Animais , Apoptose/efeitos dos fármacos , Blastocisto/efeitos dos fármacos , Massa Celular Interna do Blastocisto/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Ciclina E/genética , Células-Tronco Embrionárias/fisiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/farmacologia , Proteínas Oncogênicas/fisiologia , Oócitos , RNA de Cadeia Dupla/análise , Suínos , Proteína X Associada a bcl-2/metabolismo
4.
Apoptosis ; 22(4): 570-584, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28176146

RESUMO

Centchroman (CC) or Ormeloxifene has been shown to induce apoptosis and cell cycle arrest in various types of cancer cells. This has, however, not been addressed for endometrial cancer cells where its (CC) mechanism of action remains unclear. This study focuses on the basis of antineoplasticity of CC by blocking the targets involved in the cell cycle, survival and apoptosis in endometrial cancer cells. Ishikawa Human Endometrial Cancer Cells were cultured under estrogen deprived medium, exposed to CC and analyzed for proliferation and apoptosis. Additionally, we also analyzed oxidative stress induced by CC. Cell viability studies confirmed the IC50 of CC in Ishikawa cells to be 20 µM after 48 h treatment. CC arrests the cells in G0/G1 phase through cyclin D1 and cyclin E mediated pathways. Phosphatidylserine externalization, nuclear morphology changes, DNA fragmentation, PARP cleavage, and alteration of Bcl-2 family protein expression clearly suggest ongoing apoptosis in the CC treated cells. Activation of caspase 3 & 9, up-regulation of AIF and inhibition of apoptosis by z-VAD-fmk clearly explains the participation of the intrinsic pathway of programmed cell death. Further, the increase of ROS, loss of MMP, inhibition of antioxidant (MnSOD, Cu/Zn-SOD and GST) and inhibition of apoptosis with L-NAC suggests CC induced oxidative stress leading to apoptosis via mitochondria mediated pathway. Therefore, CC could be a potential therapeutic agent for the treatment of Endometrial Cancer adjunct to its utility as a contraceptive and an anti-breast cancer agent.


Assuntos
Adenocarcinoma/patologia , Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Centocromano/farmacologia , Neoplasias do Endométrio/patologia , Caspases/fisiologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Ciclina D1/fisiologia , Ciclina E/fisiologia , Fragmentação do DNA/efeitos dos fármacos , Feminino , Humanos , Concentração Inibidora 50 , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas de Neoplasias/fisiologia , Oxirredução , Transporte Proteico/efeitos dos fármacos
5.
Adv Exp Med Biol ; 1042: 527-547, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29357072

RESUMO

Precise replication of genetic material and its equal distribution to daughter cells are essential to maintain genome stability. In eukaryotes, chromosome replication and segregation are temporally uncoupled, occurring in distinct intervals of the cell cycle, S and M phases, respectively. Cyclin E accumulates at the G1/S transition, where it promotes S phase entry and progression by binding to and activating CDK2. Several lines of evidence from different models indicate that cyclin E/CDK2 deregulation causes replication stress in S phase and chromosome segregation errors in M phase, leading to genomic instability and cancer. In this chapter, we will discuss the main findings that link cyclin E/CDK2 deregulation to genomic instability and the molecular mechanisms by which cyclin E/CDK2 induces replication stress and chromosome aberrations during carcinogenesis.


Assuntos
Ciclina E/genética , Ciclina E/fisiologia , Instabilidade Genômica/genética , Animais , Ciclo Celular/genética , Replicação do DNA/genética , Regulação da Expressão Gênica , Humanos , Origem de Replicação/genética
6.
Gynecol Oncol ; 143(1): 152-158, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27461360

RESUMO

Cyclin E1 (CCNE1) gene amplification occurs in approximately 20% of ovarian high grade serous carcinoma (HGSC) and is associated with chemotherapy resistance and, in some studies, overall poor prognosis. The role of cyclin E1 in inducing S phase entry relies upon its interactions with cyclin dependent kinases (CDK), specifically CDK2. Therapies to target cyclin E1-related functions have centered on CDK inhibitors and proteasome inhibitors. While many studies have helped elucidate the functions and regulatory mechanisms of cyclin E1, further research utilizing cyclin E1 as a therapeutic target in ovarian cancer is warranted. This review serves to present the scientific background describing the role and function of cyclin E1 in cancer development and progression, to distinguish cyclin E1-amplified HGSC as a unique subset of ovarian cancer deserving of further therapeutic investigation, and to provide an updated overview on the studies which have utilized cyclin E1 as a target for therapy in ovarian cancer.


Assuntos
Ciclina E/fisiologia , Cistadenocarcinoma Seroso/etiologia , Proteínas Oncogênicas/fisiologia , Neoplasias Ovarianas/etiologia , Ciclina E/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/fisiologia , Cistadenocarcinoma Seroso/terapia , Feminino , Humanos , Proteínas Oncogênicas/antagonistas & inibidores , Neoplasias Ovarianas/terapia
7.
Cancer Res ; 75(19): 4164-75, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26238784

RESUMO

The proteasome is clinically validated as a target for cancer therapeutics. However, proteasome-inhibitory agents that are cancer selective have yet to be developed. In this study, we report the identification of a safe and effective proteasome inhibitor with selective anticancer properties. We screened a chemical library constructed using a hybrid approach that incorporated a 4-piperazinylquinoline scaffold and a sulfonyl phamarcophore. From this library, we identified 7-chloro-4-(4-(2,4-dinitrophenylsulfonyl)piperazin-1-yl)quinoline (VR23) as a small molecule that potently inhibited the activities of trypsin-like proteasomes (IC50 = 1 nmol/L), chymotrypsin-like proteasomes (IC50 = 50-100 nmol/L), and caspase-like proteasomes (IC50 = 3 µmol/L). Data from molecular docking and substrate competition assays established that the primary molecular target of VR23 was ß2 of the 20S proteasome catalytic subunit. Notably, VR23 was structurally distinct from other known proteasome inhibitors and selectively killed cancer cells by apoptosis, with little effect on noncancerous cells. Mechanistic investigations showed that cancer cells exposed to VR23 underwent an abnormal centrosome amplification cycle caused by the accumulation of ubiquitinated cyclin E. In combinations with the clinically approved chymotrypsin-like proteasome inhibitor bortezomib, VR23 produced a synergistic effect in killing multiple myeloma cells, including those that were resistant to bortezomib. VR23 was effective in vivo in controlling multiple myelomas and metastatic breast cancer cells, in the latter case also enhancing the antitumor activity of paclitaxel while reducing its side effects. Overall, our results identify VR23 as a structurally novel proteasome inhibitor with desirable properties as an anticancer agent.


Assuntos
Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Centrossomo/efeitos dos fármacos , Ciclina E/fisiologia , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Proteassoma/farmacologia , Quinolinas/farmacologia , Sulfonamidas/farmacologia , Proteínas Ubiquitinadas/fisiologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/secundário , Animais , Apoptose/efeitos dos fármacos , Ligação Competitiva , Bortezomib/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Centrossomo/metabolismo , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Feminino , Humanos , Camundongos , Camundongos Nus , Modelos Moleculares , Simulação de Acoplamento Molecular , Estrutura Molecular , Mieloma Múltiplo/patologia , Proteínas de Neoplasias/fisiologia , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Paclitaxel/toxicidade , Ligação Proteica , Quinolinas/administração & dosagem , Quinolinas/química , Sulfonamidas/administração & dosagem , Sulfonamidas/química , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Clin Endocrinol Metab ; 100(7): 2557-64, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25942479

RESUMO

CONTEXT: Cushing disease, due to pituitary corticotroph tumor ACTH hypersecretion, drives excess adrenal cortisol production with adverse morbidity and mortality. Loss of glucocorticoid negative feedback on the hypothalamic-pituitary-adrenal axis leads to autonomous transcription of the corticotroph precursor hormone proopiomelanocortin (POMC), consequent ACTH overproduction, and adrenal hypercortisolism. We previously reported that R-roscovitine (CYC202, seliciclib), a 2,6,9-trisubstituted purine analog, suppresses cyclin-dependent-kinase 2/cyclin E and inhibits ACTH in mice and zebrafish. We hypothesized that intrapituitary cyclin E signaling regulates corticotroph tumor POMC transcription independently of cell cycle progression. The aim was to investigate whether R-roscovitine inhibits human ACTH in corticotroph tumors by targeting the cyclin-dependent kinase 2/cyclin E signaling pathway. METHODS: Primary cell cultures of surgically resected human corticotroph tumors were treated with or without R-roscovitine, ACTH measured by RIA and quantitative PCR, and/or Western blot analysis performed to investigate ACTH and lineage-specific transcription factors. Cyclin E and E2F transcription factor 1 (E2F1) small interfering RNA (siRNA) transfection was performed in murine corticotroph tumor AtT20 cells to elucidate mechanisms for drug action. POMC gene promoter activity in response to R-roscovitine treatment was analyzed using luciferase reporter and chromatin immunoprecipitation assays. RESULTS: R-roscovitine inhibits human corticotroph tumor POMC and Tpit/Tbx19 transcription with decreased ACTH expression. Cyclin E and E2F1 exhibit reciprocal positive regulation in corticotroph tumors. R-roscovitine disrupts E2F1 binding to the POMC gene promoter and suppresses Tpit/Tbx19 and other lineage-specific POMC transcription cofactors via E2F1-dependent and -independent pathways. CONCLUSION: R-roscovitine inhibits human pituitary corticotroph tumor ACTH by targeting the cyclin E/E2F1 pathway. Pituitary cyclin E/E2F1 signaling is a previously unappreciated molecular mechanism underlying neuroendocrine regulation of the hypothalamic-pituitary-adrenal axis, providing a subcellular therapeutic target for small molecule cyclin-dependent kinase 2 inhibitors of pituitary ACTH-dependent hypercortisolism, ie, Cushing disease.


Assuntos
Antineoplásicos/uso terapêutico , Ciclina E/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Hipersecreção Hipofisária de ACTH/tratamento farmacológico , Pró-Opiomelanocortina/genética , Purinas/uso terapêutico , Adenoma Hipofisário Secretor de ACT/complicações , Adenoma Hipofisário Secretor de ACT/tratamento farmacológico , Adenoma Hipofisário Secretor de ACT/genética , Adenoma Hipofisário Secretor de ACT/patologia , Adenoma/complicações , Adenoma/tratamento farmacológico , Adenoma/genética , Adenoma/patologia , Adolescente , Adulto , Idoso , Ciclina E/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Hipersecreção Hipofisária de ACTH/patologia , Cultura Primária de Células , Pró-Opiomelanocortina/metabolismo , Roscovitina , Células Tumorais Cultivadas , Adulto Jovem
9.
Int J Cancer ; 136(6): 1361-70, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25131797

RESUMO

Apart from regulating stem cell self-renewal, embryonic development and proliferation, Bmi-1 has been recently reported to be critical in the maintenance of genome integrity. In searching for novel mechanisms underlying the anticlastogenic function of Bmi-1, we observed, for the first time, that Bmi-1 positively regulates p21 expression. We extended the finding that Bmi-1 deficiency induced chromosome breaks in multiple cancer cell models. Interestingly, we further demonstrated that knockdown of cyclin E or ectopic overexpression of p21 rescued Bmi-1 deficiency-induced chromosome breaks. We therefore conclude that p21/cyclin E pathway is crucial in modulating the anticlastogenic function of Bmi-1. As it is well established that the overexpression of cyclin E potently induces genome instability and p21 suppresses the function of cyclin E, the novel and important implication from our findings is that Bmi-1 plays an important role in limiting genomic instability in cylin E-overexpressing cancer cells by positive regulation of p21.


Assuntos
Ciclina E/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Instabilidade Genômica , Complexo Repressor Polycomb 1/fisiologia , Transdução de Sinais/fisiologia , Linhagem Celular Tumoral , Aberrações Cromossômicas , Humanos
10.
Hepatology ; 59(2): 651-60, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23787781

RESUMO

UNLABELLED: The liver has a strong regenerative capacity. After injury, quiescent hepatocytes can reenter the mitotic cell cycle to restore tissue homeostasis. This G(0) /G(1) -S cell-cycle transition of primed hepatocytes is regulated by complexes of cyclin-dependent kinase 2 (Cdk2) with E-type cyclins (CcnE1 or CcnE2). However, single genetic ablation of either E-cyclin or Cdk2 does not affect overall liver regeneration. Here, we systematically investigated the contribution of CcnE1, CcnE2, and Cdk2 for liver regeneration after partial hepatectomy (PH) by generating corresponding double- and triple-knockout (KO) mouse mutants. We demonstrate that conditional deletion of Cdk2 alone in hepatocytes resulted in accelerated induction of CcnE1, but otherwise normal initiation of S phase in vivo and in vitro. Excessive CcnE1 did not contribute to a noncanonical kinase activity, but was located at chromatin together with components of the pre-replication complex (pre-RC), such as the minichromosome maintenance (MCM) helicase. Concomitant ablation of Cdk2 and CcnE1 in hepatocytes caused a defect in pre-RC formation and further led to dramatically impaired S-phase progression by down-regulation of cyclin A2 and cell death in vitro and substantially reduced hepatocyte proliferation and liver regeneration after PH in vivo. Similarly, combined loss of CcnE1 and CcnE2, but also the Cdk2/CcnE1/CcnE2 triple KO in liver, significantly inhibited S-phase initiation and liver mass reconstitution after PH, whereas concomitant ablation of CcnE2 and Cdk2 had no effect. CONCLUSION: In the absence of Cdk2, CcnE1 performs crucial kinase-independent functions in hepatocytes, which are capable of driving MCM loading on chromatin, cyclin A2 expression, and S-phase progression. Thus, combined inactivation of Cdk2 and CcnE1 is the minimal requirement for blocking S-phase machinery in vivo.


Assuntos
Ciclina E/deficiência , Quinase 2 Dependente de Ciclina/deficiência , Replicação do DNA/fisiologia , Hepatócitos/patologia , Hepatócitos/fisiologia , Regeneração Hepática/fisiologia , Proteínas Oncogênicas/deficiência , Animais , Apoptose/fisiologia , Ciclo Celular/fisiologia , Células Cultivadas , Cromatina/fisiologia , Ciclina E/genética , Ciclina E/fisiologia , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/fisiologia , Ciclinas/deficiência , Ciclinas/genética , Ciclinas/fisiologia , Feminino , Homeostase/fisiologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/fisiologia , Fase S/fisiologia
11.
Math Biosci ; 248: 31-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24345497

RESUMO

Hypoxia is known to influence the cell cycle by increasing the G1 phase duration or by inducing a quiescent state (arrest of cell proliferation). This entry into quiescence is a mean for the cell to escape from hypoxia-induced apoptosis. It is suggested that some cancer cells have gain the advantage over normal cells to easily enter into quiescence when environmental conditions, such as oxygen pressure, are unfavorable [43,1]. This ability contributes in the appearance of highly resistant and aggressive tumor phenotypes [2]. The HiF-1α factor is the key actor of the intracellular hypoxia pathway. As tumor cells undergo chronic hypoxic conditions, HiF-1α is present in higher level in cancer than in normal cells. Besides, it was shown that genetic mutations promoting overstabilization of HiF-1α are a feature of various types of cancers [7]. Finally, it is suggested that the intracellular level of HiF-1α can be related to the aggressiveness of the tumors [53,24,4,10]. However, up to now, mathematical models describing the G1/S transition under hypoxia, did not take into account the HiF-1α factor in the hypoxia pathway. Therefore, we propose a mathematical model of the G1/S transition under hypoxia, which explicitly integrates the HiF-1α pathway. The model reproduces the slowing down of G1 phase under moderate hypoxia, and the entry into quiescence of proliferating cells under severe hypoxia. We show how the inhibition of cyclin D by HiF-1α can induce quiescence; this result provides a theoretical explanation to the experimental observations of Wen et al. (2010) [50]. Thus, our model confirms that hypoxia-induced chemoresistance can be linked, for a part, to the negative regulation of cyclin D by HiF-1α.


Assuntos
Hipóxia Celular/fisiologia , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Modelos Biológicos , Animais , Ciclina D/fisiologia , Ciclina E/fisiologia , Humanos , Conceitos Matemáticos , Neoplasias/patologia , Neoplasias/fisiopatologia , Oxigênio/metabolismo , Fosforilação , Proteína do Retinoblastoma/metabolismo
12.
Acta Med Iran ; 51(8): 513-9, 2013 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-24026986

RESUMO

Cyclin E, HER-2 and p53, are considered as major prognostic markers in breast cancer. As they are related in patho-clinical level, we aimed to check if they have any direct interaction on expression of each other. To study the effect of cyclin E on HER-2 expression, cell lines stably overexpressing cyclin E or its low molecular weight (LMW) isoforms were generated. To understand the results of p53 silencing either alone or in combination with cyclin E overexpression, we created three different p53 stably knocked down cell lines. Protein expression was analyzed by western blot, HER-2 expression in the established cell lines were determined using SYBR green real time PCR and data analyzed by REST software. Results indicate that HER-2 expression is only downregulated following p53 silencing and none of cyclin E isoforms can alter its expression. The presence of cyclin E isoforms in p53 silenced clones also does not altered HER-2 expression. Given the fact that p53 degradation is increased by HER-2 overexpression, these data can draw a regulatory loop in which a non-mutated functional p53 and HER-2 can bidirectionally regulate the expression of these two genes. This study improves our understandings of these pathways and these proteins can be introduced either as a marker or as a target in cancer treatment.


Assuntos
Neoplasias da Mama/metabolismo , Ciclina E/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Receptor ErbB-2/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Sequência de Bases , Neoplasias da Mama/patologia , Primers do DNA , Feminino , Inativação Gênica , Humanos , Células MCF-7 , Reação em Cadeia da Polimerase , Receptor ErbB-2/genética , Proteína Supressora de Tumor p53/genética
13.
J Gastroenterol Hepatol ; 28(9): 1545-54, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23574010

RESUMO

BACKGROUND AND AIM: By array-comparative genomic hybridization, we demonstrated cyclin E as one of seven genes associated with hepatocellular carcinoma (HCC) development in Ku70 DNA repair-deficient mice. We therefore explored the hypothesis that during hepatocarcinogenesis, cyclin E kinase can overcome the inhibitory effects of p53 and establish whether abnormal miRNA(mi-R)-34, a co-regulator of cyclin E and p53, can account for their interactions as "drivers" of HCC. METHODS: Dysplastic hepatocytes (DNs) and HCCs were generated from diethylnitrosamine (DEN)-injected C57BL/6 male mice at 3-12 months. RESULTS: Cyclin E/cdk2 was barely expressed in normal liver, but was readily detected in dysplastic hepatocytes, localizing to glutathione-S transferase pi-form positive cells dissected by laser-dissection. Cyclin E kinase activity preceded cyclin D1, proliferating cell nuclear antigen expression in DNs and HCCs despite maximal p53 and p21 expression. We confirmed that cyclin E, rather than cyclin D1, is the proliferative driver in hepatocarcinogenesis by immunoprecipitation experiments demonstrating preferential binding of p21 to cyclin D1, allowing cyclin E-mediated "escape" from G1/S checkpoint. We then showed cyclin E was responsible for regulating wild-type p53 by knockdown experiments in primary HCC cells; cyclin E-knockdown increased p53 and p21, diminished anti-apoptotic Bcl-XL and reduced cell viability. Conversely, blocking p53 augmented cyclin E, Bcl-XL expression and increased proliferation. Physiological interactions between cyclin E/p53/p21 were confirmed in primary hepatocytes. miR-34a,c were upregulated in dysplastic murine, human liver and HCCs compared with normal liver, and appeared to be linked to cyclin E/p53. CONCLUSION: Upregulation of functionally active cyclin E via miR34 with loss of p53 function is associated with cell-cycle checkpoint failure increasing proliferative drive that favors hepatocarcinogenesis.


Assuntos
Transformação Celular Neoplásica/patologia , Ciclina E/fisiologia , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Hepatócitos/patologia , Neoplasias Hepáticas Experimentais/patologia , Animais , Apoptose , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Ciclina E/biossíntese , Quinase 2 Dependente de Ciclina , Dietilnitrosamina , Técnicas de Silenciamento de Genes , Hepatócitos/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Hepáticas Experimentais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/fisiologia
14.
Cold Spring Harb Perspect Biol ; 5(1): a012948, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23284048

RESUMO

Developmentally programmed polyploidy occurs by at least four different mechanisms, two of which (endoreduplication and endomitosis) involve switching from mitotic cell cycles to endocycles by the selective loss of mitotic cyclin-dependent kinase (CDK) activity and bypassing many of the processes of mitosis. Here we review the mechanisms of endoreplication, focusing on recent results from Drosophila and mice.


Assuntos
Drosophila/genética , Endorreduplicação/fisiologia , Animais , Ciclo Celular/fisiologia , Ciclina E/metabolismo , Ciclina E/fisiologia , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 2 Dependente de Ciclina/fisiologia , Replicação do DNA , Drosophila/citologia , Camundongos , Poliploidia
15.
Oncogene ; 32(32): 3744-53, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22945645

RESUMO

It has become increasingly clear that oncogenes not only provide aberrant growth signals to cells but also cause DNA damage at replication forks (replication stress), which activate the ataxia telangiectasia mutated (ATM)/p53-dependent tumor barrier. Here we studied underlying mechanisms of oncogene-induced replication stress in cells overexpressing the oncogene Cyclin E. Cyclin E overexpression is associated with increased firing of replication origins, impaired replication fork progression and DNA damage that activates RAD51-mediated recombination. By inhibiting replication initiation factors, we show that Cyclin E-induced replication slowing and DNA damage is a consequence of excessive origin firing. A significant amount of Cyclin E-induced replication slowing is due to interference between replication and transcription, which also underlies the activation of homologous recombination. Our data suggest that Cyclin E-induced replication stress is caused by deregulation of replication initiation and increased interference between replication and transcription, which results in impaired replication fork progression and DNA damage triggering the tumor barrier or cancer-promoting mutations.


Assuntos
Ciclina E/fisiologia , Replicação do DNA , Transcrição Gênica , Linhagem Celular Tumoral , Dano ao DNA , Recombinação Homóloga , Humanos , Oncogenes , Rad51 Recombinase/fisiologia
16.
Hepatology ; 56(3): 1140-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22454377

RESUMO

UNLABELLED: Liver fibrogenesis is associated with the transition of quiescent hepatocytes and hepatic stellate cells (HSCs) into the cell cycle. Exit from quiescence is controlled by E-type cyclins (cyclin E1 [CcnE1] and cyclin E2 [CcnE2]). Thus, the aim of the current study was to investigate the contribution of E-type cyclins for liver fibrosis in man and mice. Expression of CcnE1, but not of its homolog, CcnE2, was induced in fibrotic and cirrhotic livers from human patients with different etiologies and in murine wild-type (WT) livers after periodical administration of the profibrotic toxin, CCl(4). To further evaluate the potential function of E-type cyclins for liver fibrogenesis, we repetitively treated constitutive CcnE1(-/-) and CcnE2(-/-) knock-out mice with CCl(4) to induce liver fibrosis. Interestingly, CcnE1(-/-) mice were protected against CCl(4)-mediated liver fibrogenesis, as evidenced by reduced collagen type I α1 expression and the lack of septum formation. In contrast, CcnE2(-/-) mice showed accelerated fibrogenesis after CCl(4) treatment. We isolated primary HSCs from WT, CcnE1(-/-), and CcnE2(-/-) mice and analyzed their activation, proliferation, and survival in vitro. CcnE1 expression in WT HSCs was maximal when they started to proliferate, but decreased after the cells transdifferentiated into myofibroblasts. CcnE1(-/-) HSCs showed dramatically impaired survival, cell-cycle arrest, and strongly reduced expression of alpha smooth muscle actin, indicating deficient HSC activation. In contrast, CcnE2-deficient HSCs expressed an elevated level of CcnE1 and showed enhanced cell-cycle activity and proliferation, compared to WT cells. CONCLUSIONS: CcnE1 and CcnE2 have antagonistic roles in liver fibrosis. CcnE1 is indispensable for the activation, proliferation, and survival of HSCs and thus promotes the synthesis of extracellular matrix and liver fibrogenesis.


Assuntos
Proliferação de Células , Ciclina E/fisiologia , Células Estreladas do Fígado/fisiologia , Cirrose Hepática/patologia , Proteínas Oncogênicas/fisiologia , Animais , Humanos , Masculino , Camundongos
17.
Oncogene ; 31(33): 3785-95, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22120721

RESUMO

The Polycomb transcription repressor BMI1 is highly expressed in human neuroblastomas and is required for the clonogenic self-renewal and tumorigenicity of human neuroblastoma cell lines. The molecular basis of BMI1 action in neuroblastoma cells is not well understood. Here we report that BMI1 has a critical role in stabilizing cyclin E1 by repressing the expression of FBXW7, a substrate-recognition subunit of the SCF E3 ubiquitin ligase that targets cyclin E1 for degradation. BMI1 binds to the FBXW7 locus in vivo and represses its mRNA expression. Overexpression of cyclin E1 or abrogation of FBXW7 induction rescues the cell-death phenotype of BMI1 knockdown. Moreover, MYCN, an oncoprotein in the pathogenesis of high-risk neuroblastomas, is able to counteract the death-inducing effect of BMI1 knockdown by activating CCNE1 transcription. We further show that high cyclin E1 expression is associated with Stage 4 neuroblastomas and poor prognosis in patients. These findings suggest a molecular mechanism for the oncogenic activity of BMI1 and MYCN in neuroblastoma pathogenesis and progression by maintaining cyclin E1 levels.


Assuntos
Biomarcadores Tumorais/fisiologia , Ciclina E/fisiologia , Neuroblastoma/mortalidade , Proteínas Nucleares/fisiologia , Proteínas Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Repressoras/fisiologia , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Ciclina E/análise , Ciclina E/genética , Inibidor p16 de Quinase Dependente de Ciclina , Progressão da Doença , Proteínas F-Box/genética , Proteína 7 com Repetições F-Box-WD , Humanos , Proteína Proto-Oncogênica N-Myc , Proteínas de Neoplasias/fisiologia , Proteínas Oncogênicas/análise , Proteínas Oncogênicas/genética , Complexo Repressor Polycomb 1 , Prognóstico , Proteína Supressora de Tumor p14ARF/fisiologia , Ubiquitina-Proteína Ligases/genética
18.
Mol Cell ; 43(3): 406-17, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21816347

RESUMO

Cyclin-dependent kinases comprise the conserved machinery that drives progress through the cell cycle, but how they do this in mammalian cells is still unclear. To identify the mechanisms by which cyclin-cdks control the cell cycle, we performed a time-resolved analysis of the in vivo interactors of cyclins E1, A2, and B1 by quantitative mass spectrometry. This global analysis of context-dependent protein interactions reveals the temporal dynamics of cyclin function in which networks of cyclin-cdk interactions vary according to the type of cyclin and cell-cycle stage. Our results explain the temporal specificity of the cell-cycle machinery, thereby providing a biochemical mechanism for the genetic requirement for multiple cyclins in vivo and reveal how the actions of specific cyclins are coordinated to control the cell cycle. Furthermore, we identify key substrates (Wee1 and c15orf42/Sld3) that reveal how cyclin A is able to promote both DNA replication and mitosis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Ciclina A2/metabolismo , Ciclina B1/metabolismo , Quinases Ciclina-Dependentes/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Sequência de Aminoácidos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular , Ciclina A2/química , Ciclina A2/fisiologia , Ciclina B1/química , Ciclina B1/fisiologia , Ciclina E/química , Ciclina E/metabolismo , Ciclina E/fisiologia , Quinases Ciclina-Dependentes/química , Quinases Ciclina-Dependentes/metabolismo , Replicação do DNA , Células HeLa , Humanos , Imunoprecipitação , Espectrometria de Massas , Dados de Sequência Molecular , Proteínas Oncogênicas/química , Proteínas Oncogênicas/metabolismo , Proteínas Oncogênicas/fisiologia , Fosforilação , Proteômica/métodos , Alinhamento de Sequência , Especificidade por Substrato
19.
Cell Cycle ; 10(2): 199-205, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21217199

RESUMO

Recent identification of the modular CLS motifs responsible for cyclins A and E localization on centrosomes has revealed a tight linkage between the nuclear and centrosomal cycles. These G1/S cyclins must localize on the centrosome in order for DNA replication to occur in the nucleus, whereas essential DNA replication factors also function on the centrosome to prevent centrosome overduplication. Both events are dependent on the presence of an intact CLS within each cyclin. Here we compare the cyclins A and E CLSs at the structural and functional levels and identify a new cyclin A CLS mutant that disrupts all CLS functions and reduces the affinity of cyclin A for Cdk2. Analysis of interactions of the CLS motif within the cyclin molecules highlights the importance of the cyclin CBOX1 region for Cdk2 binding.


Assuntos
Centrossomo/metabolismo , Ciclina A/análise , Ciclina E/análise , Sequência de Aminoácidos , Animais , Células CHO , Centrossomo/química , Cricetinae , Cricetulus , Ciclina A/metabolismo , Ciclina A/fisiologia , Ciclina E/metabolismo , Ciclina E/fisiologia , Quinase 2 Dependente de Ciclina/metabolismo , Fase G1 , Dados de Sequência Molecular , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Fase S
20.
Mol Cell Endocrinol ; 326(1-2): 25-9, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20347931

RESUMO

Pituitary tumours are benign neoplasms that may cause major endocrine dysfunction. Transgenic disruption of the cell cycle machinery frequently leads to pituitary adenoma formation in animal models. The molecular analysis of human pituitary tumours has found various alterations in the expression of cell cycle regulators: cyclins, cyclin-dependent kinases and their inhibitors. There are also different mechanisms (e.g. hypermethylation, frameshift mutations, increased proteasome degradation) responsible for changed expression in cyclin mRNA and protein. It is probable that the primary initiating events lie beyond the cell cycle and may be related to co-activation of Akt, MAP-kinase and beta-catenin pathways. Nevertheless, molecular CDK inhibitors may play a role in pituitary tumour treatment in the future.


Assuntos
Ciclinas/fisiologia , Neoplasias Hipofisárias/etiologia , Proteínas de Ciclo Celular/metabolismo , Ciclina E/metabolismo , Ciclina E/fisiologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/genética , Ciclinas/metabolismo , Humanos , Neoplasia Endócrina Múltipla Tipo 1/genética , Neoplasias , Hipófise/metabolismo , Hipófise/patologia , Neoplasias Hipofisárias/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA