Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
J Biol Chem ; 300(4): 107209, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38519029

RESUMO

FOXO1 is a transcription factor and potential tumor suppressor that is negatively regulated downstream of PI3K-PKB/AKT signaling. Paradoxically, FOXO also promotes tumor growth, but the detailed mechanisms behind this role of FOXO are not fully understood. In this study, we revealed a molecular cascade by which the Thr24 residue of FOXO1 is phosphorylated by AKT and is dephosphorylated by calcineurin, which is a Ca2+-dependent protein phosphatase. Curiously, single nucleotide somatic mutations of FOXO1 in cancer occur frequently at and near Thr24. Using a calcineurin inhibitor and shRNA directed against calcineurin, we revealed that calcineurin-mediated dephosphorylation of Thr24 regulates FOXO1 protein stability. We also found that FOXO1 binds to the promoter region of MDM2 and activates transcription, which in turn promotes MDM2-mediated ubiquitination and degradation of p53. FOXO3a and FOXO4 are shown to control p53 activity; however, the significance of FOXO1 in p53 regulation remains largely unknown. Supporting this notion, FOXO1 depletion increased p53 and p21 protein levels in association with the inhibition of cell proliferation. Taken together, these results indicate that FOXO1 is stabilized by calcineurin-mediated dephosphorylation and that FOXO1 supports cancer cell proliferation by promoting MDM2 transcription and subsequent p53 degradation.


Assuntos
Calcineurina , Proliferação de Células , Proteína Forkhead Box O1 , Proteólise , Proteínas Proto-Oncogênicas c-mdm2 , Proteína Supressora de Tumor p53 , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Humanos , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Forkhead Box O1/metabolismo , Proteína Forkhead Box O1/genética , Calcineurina/metabolismo , Calcineurina/genética , Fosforilação , Ubiquitinação , Linhagem Celular Tumoral , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/genética , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Estabilidade Proteica
2.
Proc Natl Acad Sci U S A ; 119(15): e2110256119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35394865

RESUMO

Estrogen receptor α (ERα) is a transcription factor that induces cell proliferation and exhibits increased expression in a large subset of breast cancers. The molecular mechanisms underlying the up-regulation of ERα activity, however, remain poorly understood. We identified FK506-binding protein 52 (FKBP52) as a factor associated with poor prognosis of individuals with ERα-positive breast cancer. We found that FKBP52 interacts with breast cancer susceptibility gene 1 and stabilizes ERα, and is essential for breast cancer cell proliferation. FKBP52 depletion resulted in decreased ERα expression and proliferation in breast cancer cell lines, including MCF7-derived fulvestrant resistance (MFR) cells, suggesting that inhibiting FKBP52 may provide a therapeutic effect for endocrine therapy­resistant breast cancer. In contrast, FKBP51, a closely related molecule to FKBP52, reduced the stability of ERα. Consistent with these findings, FKBP51 was more abundantly expressed in normal tissues than in cancer cells, suggesting that these FKBPs may function in the opposite direction. Collectively, our study shows that FKBP52 and FKBP51 regulate ERα stability in a reciprocal manner and reveals a regulatory mechanism by which the expression of ERα is controlled.


Assuntos
Neoplasias da Mama , Receptor alfa de Estrogênio , Proteínas de Ligação a Tacrolimo , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Estabilidade Proteica , Proteínas de Ligação a Tacrolimo/metabolismo
3.
Bioessays ; 44(12): e2200148, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36192154

RESUMO

Estrogen receptor α (ERα) is a ligand-dependent transcription factor that regulates the expression of estrogen-responsive genes. Approximately 70% of patients with breast cancer are ERα positive. Estrogen stimulates cancer cell proliferation and contributes to tumor progression. Endocrine therapies, which suppress the ERα signaling pathway, significantly improve the prognosis of patients with breast cancer. However, the development of de novo or acquired endocrine therapy resistance remains a barrier to breast cancer treatment. Therefore, understanding the regulatory mechanisms of ERα is essential to overcome the resistance to treatment. This review focuses on the regulation of ERα expression, including copy number variation, epigenetic regulation, transcriptional regulation, and stability, as well as functions from the point of view post-translational modifications.


Assuntos
Neoplasias da Mama , Receptor alfa de Estrogênio , Humanos , Feminino , Receptor alfa de Estrogênio/genética , Variações do Número de Cópias de DNA , Epigênese Genética , Neoplasias da Mama/genética , Estrogênios
4.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34711683

RESUMO

Estrogen receptor α (ER-α) mediates estrogen-dependent cancer progression and is expressed in most breast cancer cells. However, the molecular mechanisms underlying the regulation of the cellular abundance and activity of ER-α remain unclear. We here show that the protein phosphatase calcineurin regulates both ER-α stability and activity in human breast cancer cells. Calcineurin depletion or inhibition down-regulated the abundance of ER-α by promoting its polyubiquitination and degradation. Calcineurin inhibition also promoted the binding of ER-α to the E3 ubiquitin ligase E6AP, and calcineurin mediated the dephosphorylation of ER-α at Ser294 in vitro. Moreover, the ER-α (S294A) mutant was more stable and activated the expression of ER-α target genes to a greater extent compared with the wild-type protein, whereas the extents of its interaction with E6AP and polyubiquitination were attenuated. These results suggest that the phosphorylation of ER-α at Ser294 promotes its binding to E6AP and consequent degradation. Calcineurin was also found to be required for the phosphorylation of ER-α at Ser118 by mechanistic target of rapamycin complex 1 and the consequent activation of ER-α in response to ß-estradiol treatment. Our study thus indicates that calcineurin controls both the stability and activity of ER-α by regulating its phosphorylation at Ser294 and Ser118 Finally, the expression of the calcineurin A-α gene (PPP3CA) was associated with poor prognosis in ER-α-positive breast cancer patients treated with tamoxifen or other endocrine therapeutic agents. Calcineurin is thus a promising target for the development of therapies for ER-α-positive breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Calcineurina/metabolismo , Receptor alfa de Estrogênio/metabolismo , Calcineurina/fisiologia , Linhagem Celular Tumoral , Estradiol/farmacologia , Receptor alfa de Estrogênio/efeitos dos fármacos , Estrogênios/metabolismo , Feminino , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/efeitos dos fármacos
5.
Cancer Sci ; 114(7): 2729-2738, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37026526

RESUMO

FK506 binding protein 52 (FKBP52) (gene name FKBP4) is a 52 kDa protein that belongs to the FKBP family; it binds to the immunosuppressant FK506 and has proline isomerase activity. In addition to its FK domain-containing peptidylprolyl isomerase activity, FKBP52 also acts as a cochaperone through the tetratricopeptide repeat domain that mediates binding to heat shock protein 90. Previous studies have reported that FKBP52 is associated with hormone-dependent, stress-related, and neurodegenerative diseases, revealing its diverse functions. In particular, the effects of FKBP52 on cancer have attracted considerable attention. FKBP52 promotes the growth of hormone-dependent cancers by activating steroid hormone receptors. Recent studies have shown that the expression of FKBP52 is increased not only in steroid hormone-dependent cancer cells but also in colorectal, lung, and liver cancers, revealing its diverse functions that contribute to cancer growth. This review summarizes reports related to hormone-dependent cancer and cell proliferation in terms of the structure of FKBP52 and its function on interacting molecules.


Assuntos
Neoplasias Hormônio-Dependentes , Proteínas de Ligação a Tacrolimo , Humanos , Proliferação de Células/genética , Proteínas de Choque Térmico HSP90/metabolismo , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Ligação Proteica , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/química , Proteínas de Ligação a Tacrolimo/metabolismo
6.
Biochem Biophys Res Commun ; 641: 84-92, 2023 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-36525928

RESUMO

The epidermal growth factor receptor (EGFR) is highly expressed or abnormally activated in several types of cancers, such as lung and colorectal cancers. Inhibitors that suppress the tyrosine kinase activity of EGFR have been used in the treatment of lung cancer. However, resistance to these inhibitors has become an issue in cancer treatment, and the development of new therapies that inhibit EGFR is desired. We found that calcineurin, a Ca2+/calmodulin-activated serine/threonine phosphatase, is a novel regulator of EGFR. Inhibition of calcineurin by FK506 treatment or calcineurin depletion promoted EGFR degradation in cancer cells. In addition, we found that calcineurin dephosphorylates EGFR at serine (S)1046/1047, which in turn stabilizes EGFR. Furthermore, in human colon cancer cells transplanted into mice, the inhibition of calcineurin by FK506 decreased EGFR expression. These results indicate that calcineurin stabilizes EGFR by dephosphorylating S1046/1047 and promotes tumor growth. These findings suggest that calcineurin may be a new therapeutic target for cancers with high EGFR expression or activation.


Assuntos
Calcineurina , Tacrolimo , Humanos , Animais , Camundongos , Calcineurina/metabolismo , Tacrolimo/farmacologia , Serina/metabolismo , Receptores ErbB/metabolismo , Fosforilação
7.
Cell ; 132(2): 221-32, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18243098

RESUMO

DNA damage results in activation or suppression of transcription of a large number of genes. Transcriptional activation has been well characterized in the context of sequence-specific DNA-bound activators, whereas mechanisms of transcriptional suppression are largely unexplored. We show here that DNA damage rapidly reduces histone H3 Threonine 11 (T11) phosphorylation. This correlates with repression of genes, including cyclin B1 and cdk1. H3-T11 phosphorylation occurs throughout the cell cycle and is Chk1 dependent in vivo. Following DNA damage, Chk1 undergoes rapid chromatin dissociation, concomitant with reduced H3-T11 phosphorylation. Furthermore, we find that loss of H3-T11 phosphorylation correlates with reduced binding of the histone acetyltransferase GCN5 at cyclin B1 and cdk1 promoters and reduced H3-K9 acetylation. We propose a mechanism for Chk1 as a histone kinase, responsible for DNA-damage-induced transcriptional repression by loss of histone acetylation.


Assuntos
Histonas/metabolismo , Proteínas Quinases/química , Proteínas Quinases/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Transcrição Gênica , Adenoviridae/genética , Animais , Células Cultivadas , Quinase 1 do Ponto de Checagem , Meios de Cultura Livres de Soro , Dano ao DNA , Embrião de Mamíferos , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Regulação da Expressão Gênica , Células HCT116 , Histonas/genética , Humanos , Camundongos , Modelos Genéticos , Fosforilação/efeitos da radiação , Proteínas Quinases/análise , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Especificidade por Substrato , Raios Ultravioleta
8.
Genes Cells ; 26(6): 447-454, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33848395

RESUMO

UHRF1 (Ubiquitin-like with PHD and ring finger domains 1) regulates DNA methylation and histone modifications and plays a key role in cell proliferation and the DNA damage response. However, the function of UHRF2, a paralog of UHRF1, in the DNA damage response remains largely unknown. Here, we show that UHRF2 is essential for maintaining cell viability after UV irradiation, as well as for the proliferation of cancer cells. UHRF2 was found to physically interact with ATR in a DNA damage-dependent manner through UHRF2's TTD domain. In addition, phosphorylation of threonine at position 1989, which is required for UV-induced activation of ATR, was impaired in cells depleted of UHRF2, suggesting that UHRF2 is essential in ATR activation. In conclusion, these results suggest a new regulatory mechanism of ATR activation mediated by UHRF2.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Raios Ultravioleta , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Quinase 1 do Ponto de Checagem/metabolismo , Dano ao DNA , Humanos , Ligação Proteica/efeitos da radiação
9.
Mol Cell ; 55(1): 73-84, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24910096

RESUMO

Senescence is a state of permanent growth arrest and is a pivotal part of the antitumorigenic barrier in vivo. Although the tumor suppressor activities of p53 and pRb family proteins are essential for the induction of senescence, molecular mechanisms by which these proteins induce senescence are still not clear. Using time-lapse live-cell imaging, we demonstrate here that normal human diploid fibroblasts (HDFs) exposed to various senescence-inducing stimuli undergo a mitosis skip before entry into permanent cell-cycle arrest. This mitosis skip is mediated by both p53-dependent premature activation of APC/C(Cdh1) and pRb family protein-dependent transcriptional suppression of mitotic regulators. Importantly, mitotic skipping is necessary and sufficient for senescence induction. p16 is only required for maintenance of senescence. Analysis of human nevi also suggested the role of mitosis skip in in vivo senescence. Our findings provide decisive evidence for the molecular basis underlying the induction and maintenance of cellular senescence.


Assuntos
Senescência Celular , Mitose/fisiologia , Pontos de Checagem do Ciclo Celular , Inibidor p16 de Quinase Dependente de Ciclina , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiologia , Imagem com Lapso de Tempo , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/fisiologia
10.
Int J Mol Sci ; 23(3)2022 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-35163061

RESUMO

Calcineurin, a calcium-dependent serine/threonine phosphatase, integrates the alterations in intracellular calcium levels into downstream signaling pathways by regulating the phosphorylation states of several targets. Intracellular Ca2+ is essential for normal cellular physiology and cell cycle progression at certain critical stages of the cell cycle. Recently, it was reported that calcineurin is activated in a variety of cancers. Given that abnormalities in calcineurin signaling can lead to malignant growth and cancer, the calcineurin signaling pathway could be a potential target for cancer treatment. For example, NFAT, a typical substrate of calcineurin, activates the genes that promote cell proliferation. Furthermore, cyclin D1 and estrogen receptors are dephosphorylated and stabilized by calcineurin, leading to cell proliferation. In this review, we focus on the cell proliferative functions and regulatory mechanisms of calcineurin and summarize the various substrates of calcineurin. We also describe recent advances regarding dysregulation of the calcineurin activity in cancer cells. We hope that this review will provide new insights into the potential role of calcineurin in cancer development.


Assuntos
Calcineurina/metabolismo , Cálcio/metabolismo , Neoplasias/metabolismo , Ciclo Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores de Transcrição NFATC/metabolismo , Fosforilação , Transdução de Sinais
11.
Cancer Sci ; 112(7): 2739-2752, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33939241

RESUMO

DNA damage induces transcriptional repression of E2F1 target genes and a reduction in histone H3-Thr11 phosphorylation (H3-pThr11 ) at E2F1 target gene promoters. Dephosphorylation of H3-pThr11 is partly mediated by Chk1 kinase and protein phosphatase 1γ (PP1γ) phosphatase. Here, we isolated NIPP1 as a regulator of PP1γ-mediated H3-pThr11 by surveying nearly 200 PP1 interactor proteins. We found that NIPP1 inhibits PP1γ-mediated dephosphorylation of H3-pThr11 both in vivo and in vitro. By generating NIPP1-depleted cells, we showed that NIPP1 is required for cell proliferation and the expression of E2F1 target genes. Upon DNA damage, activated protein kinase A (PKA) phosphorylated the NIPP1-Ser199 residue, adjacent to the PP1 binding motif (RVxF), and triggered the dissociation of NIPP1 from PP1γ, leading to the activation of PP1γ. Furthermore, the inhibition of PKA activity led to the activation of E2F target genes. Statistical analysis confirmed that the expression of NIPP1 was positively correlated with E2F target genes. Taken together, these findings demonstrate that the PP1 regulatory subunit NIPP1 modulates E2F1 target genes by linking PKA and PP1γ during DNA damage.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dano ao DNA , Fator de Transcrição E2F1/genética , Endorribonucleases/metabolismo , Histonas/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteína Fosfatase 1/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sistemas CRISPR-Cas , Proliferação de Células , Células Cultivadas , Quinase 1 do Ponto de Checagem/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Endorribonucleases/deficiência , Endorribonucleases/isolamento & purificação , Repressão Epigenética , Regulação da Expressão Gênica , Humanos , Fosfoproteínas Fosfatases/deficiência , Fosfoproteínas Fosfatases/isolamento & purificação , Fosforilação , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/isolamento & purificação , Receptores de Neuropeptídeo Y/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Raios Ultravioleta
12.
Genes Dev ; 24(4): 333-8, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20159953

RESUMO

A balanced deoxyribonucleotide (dNTP) supply is essential for DNA repair. Here, we found that ribonucleotide reductase (RNR) subunits RRM1 and RRM2 accumulated very rapidly at damage sites. RRM1 bound physically to Tip60. Chromatin immunoprecipitation analyses of cells with an I-SceI cassette revealed that RRM1 bound to a damage site in a Tip60-dependent manner. Active RRM1 mutants lacking Tip60 binding failed to rescue an impaired DNA repair in RRM1-depleted G1-phase cells. Inhibition of RNR recruitment by an RRM1 C-terminal fragment sensitized cells to DNA damage. We propose that Tip60-dependent recruitment of RNR plays an essential role in dNTP supply for DNA repair.


Assuntos
Dano ao DNA/fisiologia , Fase G1/fisiologia , Histona Acetiltransferases/metabolismo , Ribonucleotídeo Redutases/metabolismo , Animais , Técnicas de Silenciamento de Genes , Células HeLa , Histona Acetiltransferases/genética , Humanos , Lisina Acetiltransferase 5 , Camundongos , Transativadores
13.
Biochem Biophys Res Commun ; 469(4): 960-6, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26721438

RESUMO

The mammalian maintenance methyltransferase DNMT1 [DNA (cytosine-5-)-methyltransferase 1] mediates the inheritance of the DNA methylation pattern during replication. Previous studies have shown that depletion of DNMT1 causes a severe growth defect and apoptosis in differentiated cells. However, the detailed mechanisms behind this phenomenon remain poorly understood. Here we show that conditional ablation of Dnmt1 in murine embryonic fibroblasts (MEFs) resulted in an aberrant DNA replication program showing an accumulation of late-S phase replication and causing severely defective growth. Furthermore, we found that the catalytic activity and replication focus targeting sequence of DNMT1 are required for a proper DNA replication program. Taken together, our findings suggest that the maintenance of DNA methylation by DNMT1 plays a critical role in proper regulation of DNA replication in mammalian cells.


Assuntos
Dano ao DNA/genética , Metilação de DNA/genética , Replicação do DNA/genética , Proteínas de Manutenção de Minicromossomo/genética , Origem de Replicação/genética , Proteínas Repressoras/genética , Animais , Proliferação de Células/genética , Células Cultivadas , Fibroblastos/fisiologia , Genes cdc/genética , Camundongos
14.
J Biol Chem ; 289(17): 11656-11666, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24584930

RESUMO

Histone variants play specific roles in maintenance and regulation of chromatin structures. H2ABbd, an H2A variant, possesses a highly divergent structure compared with canonical H2A and is highly expressed in postmeiotic germ cells, but its functions in the regulation of gene expression are largely unknown. In the present study, we investigated the cellular phenotype associated with enforced H2ABbd expression. Among H2A variants, H2ABbd specifically caused growth defect in human cells and induced apoptosis. H2ABbd expression resulted in degradation of inhibitor of κB-α and translocation of NF-κB into nuclei, indicating the activation of NF-κB. Intriguingly, NF-κB activity was essential for H2ABbd-induced apoptosis. H2ABbd overexpression resulted in DNA damage after release from G1/S, progressed through the S phase slowly, and induced apoptosis. Furthermore, gene expression microarray analysis revealed that expression of H2ABbd activates groups of genes involved in apoptosis and postmeiotic germ cell development, suggesting that H2ABbd might influence transcription. Taken together, our data suggest that H2ABbd may contribute to specific chromatin structures and promote NF-κB activation, which could in turn induce apoptosis in mammalian cells.


Assuntos
Apoptose/fisiologia , Histonas/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Replicação do DNA , Histonas/genética , Humanos , Reação em Cadeia da Polimerase em Tempo Real
15.
Biochem Biophys Res Commun ; 458(3): 470-475, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25660450

RESUMO

Epigenetic modifications such as DNA methylation and histone H3 lysine 27 methylation (H3K27me) are repressive marks that silence gene expression. The M phase phosphoprotein (MPP8) associates with proteins involved in both DNA methylation and histone modifications, and therefore, is a potential candidate to mediate crosstalk between repressive epigenetic pathways. Here, by performing immunohistochemical analyses we demonstrate that MPP8 is expressed in the rodent testis, especially in spermatocytes, suggesting a role in spermatogenesis. Interestingly, we found that MPP8 physically interacts with PRC1 (Polycomb Repressive Complex 1) components which are known to possess essential function in testis development by modulating monoubiquitination of Histone H2A (uH2A) and trimethylation of Histone H3 Lysine 27 (H3K27me3) residues. Knockdown analysis of MPP8 in HeLa cells resulted in derepression of a set of genes that are normally expressed in spermatogonia, spermatids and mature sperm, thereby indicating a role for this molecule in silencing testis-related genes in somatic cells. In addition, depletion of MPP8 in murine ES cells specifically induced expression of genes involved in mesoderm differentiation, such as Cdx2 and Brachyury even in the presence of LIF, which implicated that MPP8 might be required to repress differentiation associated genes during early development. Taken together, our results indicate that MPP8 could have a role for silencing genes that are associated with differentiation of the testis and the mesoderm by interacting with epigenetic repressors modules such as the PRC1 complex.


Assuntos
Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Espermatogênese , Animais , Linhagem Celular , Metilação de DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Técnicas de Silenciamento de Genes , Células HeLa , Histonas/metabolismo , Humanos , Masculino , Camundongos , Fosfoproteínas/análise , Complexo Repressor Polycomb 1/análise , Mapas de Interação de Proteínas , Ratos Endogâmicos F344 , Espermatócitos/citologia , Espermatócitos/metabolismo , Ativação Transcricional
16.
EMBO J ; 29(20): 3558-70, 2010 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-20834228

RESUMO

Although the linkage of Chk1 and Chk2 to important cancer signalling suggests that these kinases have functions as tumour suppressors, neither Chk1+/- nor Chk2-/- mice show a predisposition to cancer under unperturbed conditions. We show here that Chk1+/-Chk2-/- and Chk1+/-Chk2+/- mice have a progressive cancer-prone phenotype. Deletion of a single Chk1 allele compromises G2/M checkpoint function that is not further affected by Chk2 depletion, whereas Chk1 and Chk2 cooperatively affect G1/S and intra-S phase checkpoints. Either or both of the kinases are required for DNA repair depending on the type of DNA damage. Mouse embryonic fibroblasts from the double-mutant mice showed a higher level of p53 with spontaneous DNA damage under unperturbed conditions, but failed to phosphorylate p53 at S23 and further induce p53 expression upon additional DNA damage. Neither Chk1 nor Chk2 is apparently essential for p53- or Rb-dependent oncogene-induced senescence. Our results suggest that the double Chk mutation leads to a high level of spontaneous DNA damage, but fails to eliminate cells with damaged DNA, which may ultimately increase cancer susceptibility independently of senescence.


Assuntos
Ciclo Celular/fisiologia , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Apoptose/fisiologia , Células Cultivadas , Senescência Celular , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Aberrações Cromossômicas , Dano ao DNA , Reparo do DNA , Feminino , Fibroblastos/citologia , Fibroblastos/fisiologia , Deleção de Genes , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/patologia , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases/genética
17.
J Biochem ; 175(3): 235-244, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38030387

RESUMO

The transcription factor NFAT plays key roles in multiple biological activities, such as immune responses, tissue development and malignant transformation. NFAT is dephosphorylated by calcineurin, which is activated by intracellular calcium levels, and translocated into the nucleus, resulting in transcriptional activation. Calcineurin dephosphorylates various target proteins and regulates their functions. However, the regulation of NFAT degradation is largely unknown, and it is unclear whether calcineurin contributes to the stability of NFAT. We investigated the effect of calcineurin inhibition on NFAT protein stability and found that the dephosphorylation of NFAT by calcineurin promotes the NFAT stabilization, whereas calcineurin mutant that is defective in phosphatase activity was unable to stabilize NFAT. Increased intracellular calcium ion concentration, which is essential for calcineurin activation, also induced NFAT stability. In addition, we identified S-phase kinase associated protein 2 (Skp2), an F-box protein of the SCF ubiquitin ligase complex, as a factor mediating degradation of NFAT when calcineurin was depleted. In summary, these findings revealed that the dephosphorylation of NFAT by calcineurin protects NFAT from degradation by Skp2 and promotes its protein stability.


Assuntos
Calcineurina , Fatores de Transcrição NFATC , Calcineurina/metabolismo , Fatores de Transcrição NFATC/metabolismo , Cálcio/metabolismo , Proteínas Quinases Associadas a Fase S , Proteínas/metabolismo
18.
Sci Rep ; 14(1): 14732, 2024 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926604

RESUMO

Excess amounts of histones in the cell induce mitotic chromosome loss and genomic instability, and are therefore detrimental to cell survival. In yeast, excess histones are degraded by the proteasome mediated via the DNA damage response factor Rad53. Histone expression, therefore, is tightly regulated at the protein level. Our understanding of the transcriptional regulation of histone genes is far from complete. In this study, we found that calcineurin inhibitor treatment increased histone protein levels, and that the transcription factor NFATc1 (nuclear factor of activated T cells 1) repressed histone transcription and acts downstream of the calcineurin. We further revealed that NFATc1 binds to the promoter regions of many histone genes and that histone transcription is downregulated in a manner dependent on intracellular calcium levels. Indeed, overexpression of histone H3 markedly inhibited cell proliferation. Taken together, these findings suggest that NFATc1 prevents the detrimental effects of histone H3 accumulation by inhibiting expression of histone at the transcriptional level.


Assuntos
Calcineurina , Histonas , Fatores de Transcrição NFATC , Fatores de Transcrição NFATC/metabolismo , Fatores de Transcrição NFATC/genética , Histonas/metabolismo , Calcineurina/metabolismo , Humanos , Proliferação de Células , Regulação da Expressão Gênica , Regiões Promotoras Genéticas , Transdução de Sinais , Transcrição Gênica , Cálcio/metabolismo
19.
Life Sci Alliance ; 7(8)2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38803221

RESUMO

FK506-binding protein 52 (FKBP52) is a member of the FKBP family of proline isomerases. FKBP52 is up-regulated in various cancers and functions as a positive regulator of steroid hormone receptors. Depletion of FKBP52 is known to inhibit cell proliferation; however, the detailed mechanism remains poorly understood. In this study, we found that FKBP52 depletion decreased MDM2 transcription, leading to stabilization of p53, and suppressed cell proliferation. We identified NFATc1 and NFATc3 as transcription factors that regulate MDM2 We also found that FKBP52 associated with NFATc3 and facilitated its nuclear translocation. In addition, calcineurin, a well-known Ca2+ phosphatase essential for activation of NFAT, plays a role in MDM2 transcription. Supporting this notion, MDM2 expression was found to be regulated by intracellular Ca2+ Taken together, these findings reveal a new role of FKBP52 in promoting cell proliferation via the NFAT-MDM2-p53 axis, and indicate that inhibition of FKBP52 could be a new therapeutic tool to activate p53 and inhibit cell proliferation.


Assuntos
Proliferação de Células , Fatores de Transcrição NFATC , Proteínas Proto-Oncogênicas c-mdm2 , Proteínas de Ligação a Tacrolimo , Proteína Supressora de Tumor p53 , Humanos , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Proliferação de Células/genética , Fatores de Transcrição NFATC/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Linhagem Celular Tumoral , Cálcio/metabolismo , Calcineurina/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais
20.
Biochem Biophys Res Commun ; 432(4): 654-9, 2013 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-23416073

RESUMO

Repressive epigenetic modifications, DNA methylation at CpG sites and histone H3 lysine 9 (H3K9) methylation, are enriched in heterochromatin, which undergoes drastic changes in structure during mitosis. MPP8 (M phase phosphoprotein 8) has been proposed to regulate positive association between these two repressive modifications, but actual involvement of this protein in changes in the heterochromatin structure during mitosis remains elusive. We demonstrate here that MPP8 predominantly localized to, but dissociated from, chromatin during interphase and early mitosis, respectively. Chromatin dissociation from MPP8 appeared to correlate with the phosphorylation status of MPP8. Experiments using inhibitors of various mitotic kinases demonstrated that the chromatin dissociation of MPP8 during metaphase to anaphase was specifically regulated by cyclin B1-Cdk1. Indeed, cyclin B1-Cdk1 effectively phosphorylated MPP8 in vitro and on STA mutant of MPP8 (all possible sites phosphorylated by Cdk were substituted by alanine) failed to dissociate from chromatin during early mitosis. Taken together, our results indicate that the chromatin association of MPP8 is regulated by Cdk-dependent phosphorylation.


Assuntos
Cromatina/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Mitose , Fosfoproteínas/metabolismo , Proteína Quinase CDC2/metabolismo , Ciclina B1/metabolismo , Células HeLa , Humanos , Mutação , Fosfoproteínas/genética , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA