Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
São Paulo; s.n; s.n; 2023. 153 p. tab, graf.
Thesis in Portuguese | LILACS | ID: biblio-1437804

ABSTRACT

Proteínas tirosina-fosfatase (PTPs) possuem papel fundamental na regulação da transdução de sinais e estão envolvidas em diversos processos fundamentais do ciclo celular. As Cdc25 (Cell Division Cycle 25) são fosfatases duais encontradas em todos os organismos eucarióticos e atuam em checkpoints do ciclo celular, permitindo ou inibindo o prosseguimento deste. Este grupo de proteínas pertence à classe de PTPs com atividade baseada em cisteína, apresenta domínio catalítico altamente conservado assim como o motivo catalítico, P-loop. Devido sua função, as Cdc25 são consideradas possíveis alvos terapêuticos para tratamento de câncer e sua interação com pequenas moléculas e inibidores tem sido investigada de forma que análises estruturais e de ligação das Cdc25 com inibidores podem elucidar aspectos importantes do mecanismo de ação destes além de direcionar para o desenho racional de fármacos. Interações cátion-π são interações intra ou intermoleculares não-covalentes que ocorrem entre uma espécie química catiônica, como o grupo guanidino de argininas, e uma das faces de um sistema π rico em elétrons, como dos anéis indólicos de triptofanos. Apesar de pouco discutidas na literatura, quando em comparação às interações não-covalentes mais convencionais, do ponto de vista energético as interações cátion-π são tão importantes na estruturação de proteínas quanto às ligações de hidrogênio ou pontes salinas. De fato estas interações são observadas com frequência em estruturas proteicas resolvidas. O domínio catalítico da Cdc25B possui diversas argininas expostas em sua superfície e um único resíduo de triptofano localizado na região C-terminal flexível, muito próximo do sítio catalítico da proteína. A flexibilidade de proteínas ou de regiões proteicas apresenta importante papel no reconhecimento entre biomoléculas participantes de vias de sinalização e tem sido muito estudada atualmente. Aqui, simulações de dinâmica molecular, experimentos de 1H-15N HSQC RMN, ensaios de cinética de inibição e de ancoragem molecular, evidenciam a existência de contatos cátion-π transientes na superfície de um importante membro da família das Cdc25, a Cdc25B, e de sítios de interação entre inibidores testados e a proteína com destaque a sítios na proximidades do P-loop, região próxima ao C-terminal desordenado, onde se demonstra estabilidade da interação com os pequenos ligantes


Protein tyrosine phosphatase (PTPs) play a fundamental role in the regulation of signal transduction and are involved in several fundamental processes of the cell cycle. Cdc25 (Cell Division Cycle 25) are dual phosphatases found in all eukaryotic organisms and act at checkpoints of the cell cycle, allowing or inhibiting its progression. This group of proteins belongs to the class of PTPs with cysteine-based activity, presenting a highly conserved catalytic domain as well as the catalytic motif, P-loop. Due to their function, Cdc25 are considered possible therapeutic targets for cancer treatment and their interaction with small molecules and inhibitors has been investigated so that structural and binding analyzes of Cdc25 with inhibitors can elucidate important aspects of their mechanism of action besides directing to rational drug design. Cation-π interactions are non-covalent intra- or intermolecular interactions that occur between a cationic chemical species, such as the guanidino group of arginines, and one of the faces of an electron-rich system, such as the indole rings of tryptophans. Although little discussed in the literature, when compared to more conventional non-covalent interactions, from the energetic point of view, cation-π interactions are as important in the structuring of proteins as hydrogen bonds or salt bridges. In fact, these interactions are frequently observed in solved protein structures. The catalytic domain of Cdc25B has several arginines exposed on its surface and a single tryptophan residue located in the flexible C-terminal region, very close to the catalytic site of the protein. The flexibility of proteins or protein regions plays an important role in the recognition between biomolecules participating in signaling pathways and has been extensively studied today. Here, molecular dynamics simulations, 1H-15N HSQC NMR experiments, inhibition kinetics and molecular anchoring assays, evidence the existence of transient cation-π contacts on the surface of an important member of the Cdc25 family, Cdc25B, and of sites of interaction between tested inhibitors and the protein, with emphasis on sites in the vicinity of the P-loop, a region close to the disordered C-terminus, where stability of the interaction with the small ligands is demonstrated


Subject(s)
cdc25 Phosphatases/analysis , Molecular Docking Simulation/methods , Molecular Dynamics Simulation/classification
2.
Lab Invest ; 99(4): 483-498, 2019 04.
Article in English | MEDLINE | ID: mdl-30487595

ABSTRACT

Polyploid giant cancer cells (PGCCs) are key contributors to cancer heterogeneity, and the formation of PGCCs is associated with changes in the expression of cell-cycle-related proteins. This study investigated the intracellular localization and expression level of multiple cell-cycle-related proteins in PGCCs derived from BT-549 and HEY cells. In addition, the formation of PGCCs and the clinicopathological significance of cell-cycle-related proteins in human breast and ovarian cancer were examined. The expression levels of cell-cycle-related proteins, including cyclin B1, CDC25B, CDC25C, and other cell cycle phosphoproteins, including Chk2, and Aurora-A kinase, were determined using immunostaining and western blotting both in vitro and in vivo. Migration, invasion, and proliferation in control cells, cyclin B1 knockdown cells and their PGCCs following CoCl2 treatment were compared. In addition, human breast and ovarian cancer samples were collected to determine the correlation of number of PGCCs, expression of cell-cycle-related proteins, and tumor pathologic grade and metastasis. Our results confirm that cyclin B1 was localized in the cytoplasm of PGCCs and in the nuclei of their budding daughter cells. The phosphorylated proteins Chk2 and Aurora-A kinase regulated the expression and subcellular localization of cyclin B1, CDC25B, and CDC25C. The rate of positive cytoplasmic staining of cyclin B1 and positive nuclear staining of both CDC25B and CDC25C increased with increase in tumor grade and lymph node metastasis. Cell-cycle-related proteins, including cyclin B1, CDC25B, and CDC25C play an important role in regulating the formation of PGCCs. The inhibition of cyclinB1 and CoCl2 treatment significantly promoted cell proliferation, invasion, and migration abilities. The subcellular localization of these cell-cycle-related proteins was regulated by other cell cycle phosphoproteins, and was associated with pathologic grade and metastasis of tumors in cases of human breast and ovarian cancer.


Subject(s)
Breast Neoplasms , Cell Cycle Proteins/metabolism , Cyclin B1/metabolism , Ovarian Neoplasms , cdc25 Phosphatases/metabolism , Breast Neoplasms/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Cycle Proteins/analysis , Cell Line, Tumor , Cyclin B1/analysis , Female , Humans , Intracellular Space/metabolism , Neoplastic Processes , Ovarian Neoplasms/chemistry , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/physiopathology , Polyploidy , cdc25 Phosphatases/analysis
3.
Int J Radiat Biol ; 94(4): 394-402, 2018 04.
Article in English | MEDLINE | ID: mdl-29463172

ABSTRACT

PURPOSE: The purpose of this study is to systematically study the cell-cycle alterations of glioblastoma stem-like cells (GSLCs) after irradiation, possibly enriching the mechanisms of radioresistance of GSLCs. MATERIALS AND METHODS: GSLCs were enriched and identified, and then the radioresistance of GSLCs was validated by analyzing cell survival, cell proliferation, and radiation-induced apoptosis. The discrepancy of the cell-cycle distribution and expression of cell-cycle-related proteins between GSLCs and glioblastoma differentiated cells (GDCs) after irradiation was completely analyzed. RESULTS: The survival fractions and the cell viabilities of GSLCs were significantly higher than those of GDCs after irradiation. Radiation-induced apoptosis was less prominent in GSLCs than in GDCs. After irradiation with high-dose X-rays, the percentages of GDCs in G2/M phase was evidently increased. However, radiation-induced G2/M arrest occurred less frequently in GSLCs, but S-phase arrest occurred in GSLCs after irradiation with 8 Gy. Further mechanistic studies showed that the expressions levels of Cdc25c, Cdc2, and CyclinB1 in GSLCs were not apparently changed after irradiation, while those of p-ATM and p-Chk1 were sharply increased after irradiation in GSLCs. The basal level of Cdc25c expression in GSLCs was much higher than that in GDCs. CONCLUSIONS: We explored the cell-cycle alterations and cell-cycle-related proteins expression levels in GSLCs after irradiation, providing a novel mechanism of radioresistance of GSLCs.


Subject(s)
G2 Phase Cell Cycle Checkpoints/radiation effects , Glioblastoma/radiotherapy , M Phase Cell Cycle Checkpoints/radiation effects , Neoplastic Stem Cells/radiation effects , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins/physiology , Cell Line, Tumor , Checkpoint Kinase 1/physiology , Glioblastoma/pathology , Humans , Radiation Tolerance , Spheroids, Cellular , cdc25 Phosphatases/analysis
4.
Chem Biol Interact ; 219: 1-8, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-24858077

ABSTRACT

Cucurbitacin-I is a triterpenoids found in medicinal plants and have diverse pharmacological and biological activities. In this study, the antitumor effects of cucurbitacin-I on colon cancer and possible roles in apoptosis and cell cycle arrest were investigated. Treatment of SW480 cells, a human colon cancer cells, with cucurbitacin-I decreased cell viability and cell proliferation in a concentration-dependent manner. Also, cucurbitacin-I induced G2/M phase cell cycle arrest in SW480 cells with a decreased expression of cell cycle proteins including cyclin B1, cyclin A, CDK1, and CDC25C. Moreover, cucurbitacin-I induced increased cleavage of caspase-3, -7, -8, -9, and poly ADP ribose polymerase. When we examined the inhibitory effect of cucurbitacin-I on tumor growth in vivo, cucurbitacin-I effectively inhibited the tumorigenicity and growth of CT-26 cells in syngenic BALB/c mice. In summary, the present study showed that cucurbitacin-I reduced colon cancer cell proliferation by enhancing apoptosis and causing cell cycle arrest at the G2/M phase.


Subject(s)
Apoptosis/physiology , Cell Cycle Checkpoints/physiology , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Cucurbitaceae/chemistry , Triterpenes/pharmacology , Animals , Blotting, Western , CDC2 Protein Kinase , Caspases/metabolism , Cell Line, Tumor , Cyclin A/analysis , Cyclin A/metabolism , Cyclin B1/analysis , Cyclin B1/metabolism , Cyclin-Dependent Kinases/analysis , Cyclin-Dependent Kinases/metabolism , Flow Cytometry , Humans , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Triterpenes/therapeutic use , cdc25 Phosphatases/analysis , cdc25 Phosphatases/metabolism
5.
J BUON ; 18(4): 1006-11, 2013.
Article in English | MEDLINE | ID: mdl-24344030

ABSTRACT

PURPOSE: To investigate the expression of CDC25B, which is a member of the cyclin-dependent kinase activating phosphatase family, in diffuse astrocytoma (DA), anaplastic astrocytoma (AA), glioblastoma multiforme (GBM), pilocytic astrocytoma (PA) and reactive gliosis (RG). Also, to study the relationship of the expression level of CDC25B with clinical parameters and with p53 and Ki-67 proliferation index (PI). METHODS: Tissues were collected from 36 cases diagnosed with astrocytoma (10 DA, 6 AA, 20 GBM), 10 PA, 10 RG and 10 normal brain tissues for controlling purposes. The sections were immunohistochemically stained with CDC25B, Ki-67 and p53. For each marker, 1000 tumor cells were counted and the ratio of positive tumor cells was calculated. RESULTS: The average CDC2B staining index (CSI) was 0.6% in PA, 0.4% in DA , 7.7% in AA and 25.5% in GBM. The increase of CSI in parallel with the increase of WHO grade was significant (p=0.001). No expressions were identified in RG and normal brain. There was also significant relationship between the tumor size and CSI (p=0.027) and also between Ki-67 PI and CSI (p=0.001). Among the groups with low and high CSI in astrocytoma cases, the disease free survival (DFS) was significantly higher in the low CSI group (p=0.0001). CONCLUSIONS: Positive expression of CDC25B in astrocytoma affects the prognosis in an adverse manner. CSI can be used as a diagnostic method and CDC25B may be a possible target molecule for treatment.


Subject(s)
Astrocytoma/chemistry , Brain Neoplasms/chemistry , Brain/metabolism , Gliosis , Ki-67 Antigen/analysis , Tumor Suppressor Protein p53/analysis , cdc25 Phosphatases/analysis , Adolescent , Adult , Astrocytoma/pathology , Astrocytoma/therapy , Brain/pathology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Proliferation , Chi-Square Distribution , Disease-Free Survival , Female , Glioblastoma/chemistry , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Grading , Proportional Hazards Models , Risk Factors , Tumor Burden
6.
Mol Carcinog ; 50(2): 113-22, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21229608

ABSTRACT

Flavonoids are polyphenolic compounds which display a vast array of biological activities and are among the most promising anti-cancer agents. The derivative of quercetin, 5,7,3'-trihydroxy-3,4'-dimethoxyflavone (THDF), is a natural flavonoid that inhibits cell proliferation and induces apoptosis in human leukemia cells. Here we show that THDF induces cell-cycle arrest in the M phase and inhibits tubulin polymerization. This was associated with the accumulation of cyclin B1 and p21(Cip1) , changes in the phosphorylation status of cyclin B1, Cdk1, Cdc25C, and MPM-2, and activation of the acidic sphingomyelinase (ASMase). Moreover, desipramine attenuated THDF-mediated cell death, indicating a crucial role of ASMase in the mechanism of cell death. In vivo studies on the athymic nude mouse xenograft model also confirmed that THDF inhibits growth of human leukemia cells and suggest that this compound may have therapeutic value.


Subject(s)
Flavones/pharmacology , Sphingomyelins/metabolism , Tubulin Modulators/pharmacology , Animals , Antineoplastic Agents/pharmacology , CDC2 Protein Kinase/analysis , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cyclin B1/metabolism , Desipramine/pharmacology , Female , HL-60 Cells , Humans , Leukemia/drug therapy , Mice , Mice, Nude , Phosphorylation , Polymerization/drug effects , Sphingomyelin Phosphodiesterase/analysis , Tubulin/metabolism , U937 Cells , Xenograft Model Antitumor Assays , cdc25 Phosphatases/analysis
7.
Nutr Cancer ; 62(5): 611-21, 2010.
Article in English | MEDLINE | ID: mdl-20574922

ABSTRACT

Diets rich in n-3 polyunsaturated fatty acids (PUFAs) have been associated with a reduced risk of several types of cancer. Recent reports have suggested that these PUFAs enhance the cytotoxic effect of cancer chemoradiotherapy. The effect of docosahexaenoic acid (DHA) on key cell cycle regulators and target proteins of cancer therapy was investigated in the human malign colon cancer cell line SW620. Cell cycle check point proteins such as p21 and stratifin (14-3-3 sigma) increased at mRNA and protein level, whereas cell cycle progression proteins such as cell division cycle 25 homolog and cyclin-dependent kinase 1 decreased after DHA treatment. Protein levels of inhibitors of apoptosis family members associated with chemotherapy resistance and cancer malignancy, survivin and livin, decreased after the same treatment: likewise the expression of NF-kappaB. Levels of the proapoptotic proteins phosphorylated p38 MAPK and growth arrest-inducible and DNA damage-inducible gene 153/C/EBP-homologous protein (CHOP) increased. The results indicate that DHA treatment causes simultaneous cell cycle arrest in both the G1 and G2 phase. In conclusion, DHA affects several target proteins of chemotherapy in a favorable way. This may explain the observed enhanced chemosensitivity in cancer cells supplemented with n-3 PUFAs and encourage further studies investigating the role of n-3 PUFAs as adjuvant to chemotherapy and radiotherapy in vivo.


Subject(s)
Colonic Neoplasms/drug therapy , Docosahexaenoic Acids/pharmacology , Apoptosis , CDC2 Protein Kinase/analysis , Cell Line, Tumor , Colonic Neoplasms/chemistry , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm , G1 Phase/drug effects , G2 Phase/drug effects , Humans , Transcription Factor RelA/analysis , cdc25 Phosphatases/analysis , p38 Mitogen-Activated Protein Kinases/analysis
8.
Acta Pharmacol Sin ; 30(4): 442-50, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19305423

ABSTRACT

AIM: The aim of this study was to investigate the mechanism of pseudolaric acid B (PLAB)-induced cell cycle arrest in human melanoma SK-28 cells. METHODS: Cell growth inhibition was detected by MTT assay, the cell cycle was analyzed by flow cytometry, and protein expression was examined by Western blot analysis. RESULTS: PLAB inhibited the growth of human melanoma cells and induced G(2)/M arrest in SK-28 cells, accompanied by an up-regulation of Cdc2 phosphorylation and a subsequent down-regulation of Cdc2 expression. Furthermore, PLAB decreased the expression of Cdc25C phosphatase and increased the expression of Wee1 kinase. Meanwhile, a reduction in Cdc2 activity was partly due to induction of the expression of p21(waf1/cip1) in a p53-dependent manner. In addition, PLAB activated the checkpoint kinase, Chk2, and increased the expression of p53, two major targets of ATM kinase. These effects were inhibited by caffeine, an ATM kinase inhibitor. We also found that PLAB significantly enhanced ATM kinase activity. CONCLUSION: Taken together, these results suggest that PLAB induced G(2)/M arrest in human melanoma cells via a mechanism involving the activation of ATM, and the effect of PLAB on Cdc2 activity was mediated via interactions with the Chk2-Cdc25C and p53 signalling pathways, two distinct downstream pathways of ATM. PLAB may be a promising chemopreventive agent for treating human melanoma.


Subject(s)
Cell Cycle Proteins/physiology , Cell Division/drug effects , DNA-Binding Proteins/physiology , Diterpenes/pharmacology , Drugs, Chinese Herbal/pharmacology , G2 Phase/drug effects , Protein Serine-Threonine Kinases/physiology , Signal Transduction/drug effects , Tumor Suppressor Proteins/physiology , Ataxia Telangiectasia Mutated Proteins , CDC2 Protein Kinase , Caffeine/pharmacology , Cell Cycle Proteins/analysis , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 2 , Cyclin B/analysis , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21/analysis , Cyclin-Dependent Kinases , Humans , Melanoma/pathology , Nuclear Proteins/analysis , Protein-Tyrosine Kinases/analysis , Tumor Suppressor Protein p53/analysis , cdc25 Phosphatases/analysis
9.
Cancer Lett ; 278(2): 192-200, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19217710

ABSTRACT

We describe the events triggered by a sub-lethal concentration of airborne particulate matter (PM(10)) in A549 cells, which include the formation DNA double-strand breaks, gammaH2A.X generation, and 53BP1 recruitment. To protect the genome, cells activated ATM/ATR/Chk1/Chk2/p53 pathway but, after 48 h, cells turned into a senescence-like state. Trolox, an antioxidant, was able to prevent most of the alterations observed after particulate matter exposure, demonstrating the important role of ROS as mediator of PM(10)-induced genotoxicity and suggesting that DNA damage could be the mechanisms by which particulate matter augment the risk of lung cancer.


Subject(s)
DNA Damage , Particulate Matter/toxicity , Apoptosis , Ataxia Telangiectasia Mutated Proteins , Cell Cycle , Cell Cycle Proteins/analysis , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA-Binding Proteins/analysis , Humans , Protein Serine-Threonine Kinases/analysis , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/analysis , cdc25 Phosphatases/analysis
10.
Biochim Biophys Acta ; 1793(3): 462-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19185590

ABSTRACT

Activation of cyclin-dependent kinase complexes (CDK) at key cell cycle transitions is dependent on their dephosphorylation by CDC25 dual-specificity phosphatases (CDC25A, B and C in human). The CDC25B phosphatase plays an essential role in controlling the activity of CDK1-cyclin B complexes at the entry into mitosis and together with polo-like kinase 1 (PLK1) in regulating the resumption of cell cycle progression after DNA damage-dependent checkpoint arrest in G2. In this study, we analysed the regulation of CDC25B-dependent mitosis entry by PLK1. We demonstrate that PLK1 activity is essential for the relocation of CDC25B from the cytoplasm to the nucleus. By gain and loss of function analyses, we show that PLK1 stimulates CDC25B-induced mitotic entry in both normal conditions and after DNA-damage induced G2/M arrest. Our results support a model in which the relocalisation of CDC25B to the nucleus at the G2-M transition by PLK1 regulates its mitotic inducing activity.


Subject(s)
Cell Cycle Proteins/metabolism , Mitosis/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , cdc25 Phosphatases/metabolism , Cell Division , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , DNA Damage , Fluorescent Antibody Technique , G2 Phase , Humans , Transfection , cdc25 Phosphatases/analysis , Polo-Like Kinase 1
11.
Nutr Cancer ; 60 Suppl 1: 51-60, 2008.
Article in English | MEDLINE | ID: mdl-19003581

ABSTRACT

Withaferin A (WA) is derived from the medicinal plant Withania somnifera that has been safely used for centuries in the Indian Ayurvedic medicine for treatment of various ailments. We now demonstrate that WA treatment causes G2 and mitotic arrest in human breast cancer cells. Treatment of MDA-MB-231 (estrogen-independent) and MCF-7 (estrogen-responsive) cell lines with WA resulted in a concentration- and time-dependent increase in G2-M fraction, which correlated with a decrease in levels of cyclin-dependent kinase 1 (Cdk1), cell division cycle 25C (Cdc25C) and/or Cdc25B proteins, leading to accumulation of Tyrosine15 phosphorylated (inactive) Cdk1. Ectopic expression of Cdc25C conferred partial yet significant protection against WA-mediated G2-M phase cell cycle arrest in MDA-MB-231 cells. The WA-treated MDA-MB-231 and MCF-7 cells were also arrested in mitosis as judged by fluorescence microscopy and analysis of Ser10 phosphorylated histone H3. Mitotic arrest resulting from exposure to WA was accompanied by an increase in the protein level of anaphase promoting complex/cyclosome substrate securin. In conclusion, the results of this study suggest that G2-M phase cell cycle arrest may be an important mechanism in antiproliferative effect of WA against human breast cancer cells.


Subject(s)
Breast Neoplasms/drug therapy , Cell Division/drug effects , Ergosterol/analogs & derivatives , G2 Phase/drug effects , Medicine, Ayurvedic , Breast Neoplasms/pathology , Cell Line, Tumor , Ergosterol/pharmacology , Female , Histones/metabolism , Humans , Mitosis/drug effects , Phosphorylation , Reactive Oxygen Species/metabolism , Withanolides , cdc25 Phosphatases/analysis , cdc25 Phosphatases/physiology
12.
Cancer Invest ; 26(7): 734-40, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18665474

ABSTRACT

BACKGROUND: Cell cycle progression is regulated by cyclin dependent kinases (cdk) and cdk inhibitors. Recent immunohistological studies suggested that dysregulation of cyclin A, cyclin D, cyclin E, p16(ink4), p21(waf1/cip1), and p27(kip1) are of prognostic value in patients with breast cancer. Our study represents the first comprehensive immunohistochemical cell cycle marker analysis for cdc25A, cyclin A, cyclin D, cyclin E, p16(ink4), p21(waf1/cip1), p27(kip1), and pRb in tumor tissue and adjacent benign breast tissue from 69 primarily untreated breast cancer patients. METHODS: Immunhistochemistry using primary monoclonal antibodies to detect cdc 25A, cyclin A, cyclin D, cyclin E, p16(ink4), p21(waf1/cip1), p27(kip1), and pRb has been performed. RESULTS: Sixty-nine patients with untreated, invasive breast cancer (n = 69) were divided into a low/ intermediate and a high risk group according to the St. Gallen 2005 consensus conference. High risk patients (n = 22) had a significantly (p = 0.003) shorter mean and median survival (282.85 weeks; 383.0 weeks, respectively) than low/intermediate risk patients (375.41 weeks; not reached yet, respectively). A subgroup of high risk breast cancer patients characterized in addition by overexpression of cdc25A, cyclin A, cyclin E, p16(ink4a), and p27(kip1) experienced a shortened mean survival of 222.03, 235.71, 257.25, 239.18, and 261.94 weeks, respectively. Regarding benign breast tissue adjacent to breast cancer tissue, 59.4% of the patients investigated overexpressed cdc25A, 23.2% overexpressed pRb, and 63.2% exerted dysregulation of p27(kip1) while they proved to be negative for immunohistochemical staining regarding all other markers tested. CONCLUSION: The immunohistological analyses of cdc25A, cyclin A, cyclin E, p16(ink4a), and p27(kip1) have the potential for further refining the risk assessment in patients with untreated breast cancer who belong to the high risk category defined according to the St. Gallen 2005 consensus conference. These cell cycle markers define a subgroup of high risk patients with even higher risk of metastazation and shortened survival. For confirmation a prospective study using standardized laboratory procedures in a larger population is needed.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Cell Cycle Proteins/analysis , Cell Cycle , Cell Proliferation , Adult , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cyclin A/analysis , Cyclin D , Cyclin E/analysis , Cyclin-Dependent Kinase Inhibitor p16/analysis , Cyclin-Dependent Kinase Inhibitor p21/analysis , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/analysis , Disease-Free Survival , Female , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/analysis , Kaplan-Meier Estimate , Neoplasm Invasiveness , Proportional Hazards Models , Retinoblastoma Protein/analysis , Retrospective Studies , Risk Assessment , Treatment Outcome , cdc25 Phosphatases/analysis
13.
Biochem Biophys Res Commun ; 370(3): 483-8, 2008 Jun 06.
Article in English | MEDLINE | ID: mdl-18384749

ABSTRACT

Cdc25 dual-specificity phosphatases coordinate entry into mitosis through activating dephosphorylation of the Mitosis-Promoting Factor, Cdk1-cyclin B1. Activation of Cdc25C at the G2/M transition, involves its dissociation from 14-3-3, together with its hyperphosphorylation on several sites within its regulatory N-terminal domain, mediated by cyclin-dependent kinases and Plk1. Growing evidence suggests that phosphorylation intermediates are likely to precede complete hyperphosphorylation of Cdc25C. To address whether such variants occur in mitotic cells, we raised antibodies directed against different mitotic phosphorylation sites of human Cdc25C, and characterized the phosphorylated species detectable in HeLa cells. In the present study, we provide first-time evidence for the existence of multiple species of Cdc25C in mitotic cell extracts, including full-length and splice variants with different phosphorylation patterns, thereby revealing an intricate network of Cdc25C phosphatases, likely to have distinct biological functions.


Subject(s)
Mitosis , cdc25 Phosphatases/metabolism , Alternative Splicing , Amino Acid Sequence , Antibodies, Phospho-Specific/immunology , Cell Extracts/chemistry , Electrophoresis , HeLa Cells , Humans , Interphase , Molecular Sequence Data , Phosphoproteins/analysis , Phosphorylation , cdc25 Phosphatases/analysis , cdc25 Phosphatases/genetics
14.
Dev Biol ; 317(1): 260-9, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18367163

ABSTRACT

CDK1 is a pivotal regulator of resumption of meiosis and meiotic maturation of oocytes. CDC25A/B/C are dual-specificity phosphatases and activate cyclin-dependent kinases (CDKs). Although CDC25C is not essential for either mitotic or meiotic cell cycle regulation, CDC25B is essential for CDK1 activation during resumption of meiosis. Cdc25a -/- mice are embryonic lethal and therefore a role for CDC25A in meiosis is unknown. We report that activation of CDK1 results in a maturation-associated decrease in the amount of CDC25A protein, but not Cdc25a mRNA, such that little CDC25A is present by metaphase I. In addition, expression of exogenous CDC25A overcomes cAMP-mediated maintenance of meiotic arrest. Microinjection of Gfp-Cdc25a and Gpf-Cdc25b mRNAs constructs reveals that CDC25A is exclusively localized to the nucleus prior to nuclear envelope breakdown (NEBD). In contrast, CDC25B localizes to cytoplasm in GV-intact oocytes and translocates to the nucleus shortly before NEBD. Over-expressing GFP-CDC25A, which compensates for the normal maturation-associated decrease in CDC25A, blocks meiotic maturation at MI. This MI block is characterized by defects in chromosome congression and spindle formation and a transient reduction in both CDK1 and MAPK activities. Lastly, RNAi-mediated reduction of CDC25A results in fewer oocytes resuming meiosis and reaching MII. These data demonstrate that CDC25A behaves differently during female meiosis than during mitosis, and moreover, that CDC25A has a function in resumption of meiosis, MI spindle formation and the MI-MII transition. Thus, both CDC25A and CDC25B are critical for meiotic maturation of oocytes.


Subject(s)
Meiosis , Oocytes/cytology , Oocytes/enzymology , cdc25 Phosphatases/metabolism , Animals , Cyclic AMP/metabolism , Female , Gene Expression , Mice , Oocytes/chemistry , cdc25 Phosphatases/analysis
15.
Cell Cycle ; 7(3): 401-6, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18235220

ABSTRACT

The Cyclin-Dependent Kinase (CDK)-activating phosphatase CDC25B, localises to the centrosomes where its activity is both positively and negatively regulated by several kinases including Aurora A and CHK1. Our recent data also demonstrate a role for CDC25B in the centrosome duplication cycle and microtubule nucleation in interphase that appears to involve the recruitment of gamma-tubulin to the centrosomes. In the present study, we report that CDC25B, along with CHK1, CDK1 and WEE1, localize asymmetrically around the mother centrosome from S to G2-phases, and gradually become evenly distributed to the two centrosomes by late G2 phase, concomitant with centrosome maturation. We further demonstrate that siRNA inhibition of CDC25B results in an accumulation of cells in G2 phase with two separated centrosomes, each containing only a single centriole, suggesting a requirement for CDC25B in centriole duplication. We propose that the localisation of key cell cycle regulators to the mother centrosome ensures synchrony between the centrosome duplication and cell division cycles.


Subject(s)
Centrosome/enzymology , Interphase/physiology , cdc25 Phosphatases/metabolism , Cell Division/physiology , Centrosome/physiology , HeLa Cells , Humans , cdc25 Phosphatases/analysis , cdc25 Phosphatases/genetics
16.
Reproduction ; 135(4): 431-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18296513

ABSTRACT

It is clear from a wide range of studies that the nuclear/cytoplasmic distribution of Cdc25C has important functional consequences for cell cycle control. It is now admitted that in somatic cells, the localization of Cdc25C in the cytoplasm is required to maintain the cell in an interphasic state and that Cdc25C has to translocate to the nucleus just before M-phase to induce mitotic events. We characterized the expression and localization of Cdc25C during oocyte maturation, the first embryo mitosis, and the first steps of somatic cell nuclear transfer (SCNT) in cattle. We demonstrated that Cdc25C was expressed throughout the maturation process and the early development. We clearly showed that Cdc25C was localized in the nucleus at the germinal vesicle stage and during the early development until the blastocyst stage. However, the signal change in blastocyst and Cdc25C became cytoplasmic as is the case in somatic cells. Thus, oocytes and early embryonic cells presented a specific nuclear Cdc25C localization different from the one observed in somatic cells, suggesting that Cdc25C could have a particular localization/regulation in undifferentiated cells. Following SCNT, Cdc25C became nuclear as soon as the nucleus swelled, and this localization persisted until the blastocyst stage, as is the case in in vitro fertilized embryos. The Cdc25C nuclear localization appeared to constitute a major change, which could be associated with the reorganization of the somatic nucleus upon nuclear transfer.


Subject(s)
Blastocyst/enzymology , Cell Nucleus/enzymology , Nuclear Transfer Techniques , Oocytes/enzymology , cdc25 Phosphatases/analysis , Animals , Blotting, Western , Cattle , Cytoplasm/enzymology , Female , Fertilization in Vitro , Immunohistochemistry , Microscopy, Fluorescence , Pregnancy
17.
Life Sci ; 82(5-6): 315-23, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-18191419

ABSTRACT

Dual-specific phosphatases Cdc25 play a critical role in the cell cycle regulation by activating kinases of Cdk/cyclin complexes. Three Cdc25 isoforms (A, B and C) have been identified in mammalians. Cdc25A and B display oncogenic properties and are over-expressed in different tumors. Cdc25 phosphatases are therefore attractive targets for therapeutic strategies. Novel maleic anhydride derivatives bearing a fatty acid chain of variable size have been synthesized and tested for their Cdc25 inhibitory potential using an in vitro assay. We report biological activity of ineffective, moderate, and efficient inhibitors on breast cancer cells (MCF7) and its counterpart resistant to vincristine (Vcr-R). The most potent compounds induced Cdk2 inhibition and accumulation in G0/G1 phase of the cell cycle. Moreover, apoptosis was triggered within 48-h treatment, without oxidative burst and modulation of the Bax to Bcl-2 ratio. When used as pre-treatments, these derivatives were also able to potentiate adriamycin and cisplatin toxicity in both cell lines. Thus, maleic anhydride derivatives may mediate apoptosis through a cell cycle blockage via inhibition of Cdc25. This class of inhibitors may present potential interest in therapeutic strategies against cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Maleic Anhydrides/pharmacology , cdc25 Phosphatases/antagonists & inhibitors , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Drug Therapy, Combination , Enzyme Inhibitors/chemistry , Female , G1 Phase/drug effects , Humans , Maleic Anhydrides/chemistry , Quantitative Structure-Activity Relationship , Resting Phase, Cell Cycle/drug effects , Vincristine/pharmacology , cdc25 Phosphatases/analysis
18.
Appl Immunohistochem Mol Morphol ; 15(3): 267-72, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17721270

ABSTRACT

To clarify the significance of cdc25B, which plays an important physiologic role in regulation of the G2/M check point, in progression of gastric cancer, 125 samples of paraffin-embedded gastric cancers were investigated by immunohistochemistry. In addition, 3 human gastric cancer cell lines were studied to determine the cellular localization of cdc25B by immunohistochemistry and cell fractionation followed by Western blotting. In the cell lines cdc25B was found to be present in both nuclei and cytoplasm, but predominantly in nuclei. High labeling indices of cdc25B in invasion front of gastric cancer was observed in 31 out of 125 cases (24.8%), linked to an advanced depth of cancer invasion (P=0.02), high rates of lymphatic invasion (P=0.03), and lymph node metastasis (P<0.01). Furthermore, the Kaplan-Meier method demonstrated a poor prognosis for cdc25B high labeling indices cases (P=0.02), although multivariate analysis revealed it not to be an independent factor. In conclusion, it seems likely that cdc25B is located predominantly in nuclei when overexpressed and this has some linkage with progression of gastric cancer.


Subject(s)
Cell Cycle Proteins/analysis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , cdc25 Phosphatases/analysis , Aged , Blotting, Western , Cell Fractionation , Cell Line, Tumor , Disease Progression , Female , Humans , Immunohistochemistry , Male , Multivariate Analysis , Prognosis , Stomach Neoplasms/pathology , Survival Analysis
19.
Oncogene ; 26(57): 7847-58, 2007 Dec 13.
Article in English | MEDLINE | ID: mdl-17599046

ABSTRACT

CDC25 phosphatases control cell-cycle progression by dephosphorylating and activating cyclin-dependent kinases. CDC25B, one of the three members of this family in human cells, is thought to regulate initial mitotic events. CDC25B is an unstable protein whose proteasomal degradation is proposed to be controlled by beta-TrCP. Here, we have investigated the regulation of CDC25B during mitosis, using time-lapse video microscopy. We found that CDC25B expression is high during early mitosis, and that its degradation occurs after the metaphase-anaphase transition and cyclin B1 destruction. We also show that CDC25B degradation after metaphase is dependent on the integrity of the KEN-box and RRKSE motifs that are located within the alternatively spliced B domain, and that the CDC25B2 splice variant, that lacks this domain, is stable during mitosis. Furthermore, we show that the N-terminal region of CDC25B, encompassing the B domain, undergoes major conformational changes during mitosis that can be monitored by intramolecular fluorescence resonance energy transfer variation of specific CDC25B biosensors. This study demonstrates that CDC25B splice variants have differential mitotic stabilities, a feature that is likely to have major consequences on the local control of cyclin-dependent kinase-cyclin activities during mitotic progression.


Subject(s)
Mitosis , cdc25 Phosphatases/metabolism , Animals , CDC2 Protein Kinase/metabolism , Cyclin B/metabolism , Fluorescence Resonance Energy Transfer , HeLa Cells , Humans , Metaphase , Mice , Protein Structure, Tertiary , beta-Transducin Repeat-Containing Proteins/metabolism , cdc25 Phosphatases/analysis , cdc25 Phosphatases/chemistry , cdc25 Phosphatases/genetics
20.
Virchows Arch ; 450(1): 65-71, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17111124

ABSTRACT

Cells expressing high-risk human papillomavirus (HPV) E7 protein display impaired checkpoint control after DNA damage and exhibit elevated rates of mutagenesis. Repression of HPV E7 expression results in the subsequent accumulation of hypophosphorylated retinoblastoma protein and repression of the Cdc25A genes. No study has been conducted to elucidate the role of Cdc25A in the development and progression of human oral carcinomas. To confirm Cdc25A protein expression together with HPV, immunohistochemistry, Western blotting, polymerase chain reaction (PCR), and reverse transcriptase (RT)-PCR were performed using various histological subtypes of oral carcinomas. Cdc25A protein was localized predominantly in the cell nuclei in carcinomas, and high expression was found in 54% of primary tumors. HPV-16 E7 was not found in non-neoplastic oral tissues, whereas it was observed in eight (36%) of 22 oral carcinomas. We found a significant correlation between Cdc25A over-expression and HPV-16 E7 positive carcinomas. There was a strong positive correlation between Cdc25A over-expression and tumor size and TNM stage. This study suggests that Cdc25A is likely to be an important mediator in the progression of oral tumors, and HPV-16 E7 may be a sensitive indicator of the involvement of viral oncogenes in oral carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/virology , Mouth Neoplasms/virology , Oncogene Proteins, Viral/genetics , cdc25 Phosphatases/analysis , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/pathology , DNA, Viral/analysis , Female , Humans , Immunohistochemistry , Male , Middle Aged , Mouth Neoplasms/chemistry , Mouth Neoplasms/pathology , Papillomavirus E7 Proteins , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Risk
SELECTION OF CITATIONS
SEARCH DETAIL
...