Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(3)2022 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-35163809

RESUMO

Long non-coding RNAs (lncRNAs) play important biological roles. Here, the roles of the lncRNA KCNQ1OT1 in cellular senescence and calorie restriction were determined. KCNQ1OT1 knockdown mediated various senescence markers (increased senescence-associated ß-galactosidase staining, the p53-p21Cip1/WAF1 pathway, H3K9 trimethylation, and expression of the senescence-associated secretory phenotype) and reactive oxygen species generation via CK2α downregulation in human cancer HCT116 and MCF-7 cells. Additionally, KCNQ1OT1 was downregulated during replicative senescence, and its silencing induced senescence in human lung fibroblast IMR-90 cells. Additionally, an miR-760 mimic suppressed KCNQ1OT1-mediated CK2α upregulation, indicating that KCNQ1OT1 upregulated CK2α by sponging miR-760. Finally, the KCNQ1OT1-miR-760 axis was involved in both lipopolysaccharide-mediated CK2α reduction and calorie restriction (CR)-mediated CK2α induction in these cells. Therefore, for the first time, this study demonstrates that the KCNQ1OT1-miR-760-CK2α pathway plays essential roles in senescence and CR, thereby suggesting that KCNQ1OT1 is a novel therapeutic target for an alternative treatment that mimics the effects of anti-aging and CR.


Assuntos
Restrição Calórica/efeitos adversos , Fibroblastos/citologia , MicroRNAs/genética , Neoplasias/genética , Caseína Quinase II/genética , Linhagem Celular , Senescência Celular , Regulação para Baixo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , Lipopolissacarídeos/efeitos adversos , Células MCF-7 , Neoplasias/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Espécies Reativas de Oxigênio/metabolismo , Fenótipo Secretor Associado à Senescência/efeitos dos fármacos
2.
Int J Mol Sci ; 22(1)2021 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-33401686

RESUMO

Senescent cells secrete pro-inflammatory factors, and a hallmark feature of senescence is senescence-associated secretory phenotype (SASP). The aim of this study is to investigate the protein kinase CK2 (CK2) effects on SASP factors expression in cellular senescence and organism aging. Here CK2 down-regulation induced the expression of SASP factors, including interleukin (IL)-1ß, IL-6, and matrix metalloproteinase (MMP) 3, through the activation of nuclear factor-κB (NF-κB) signaling in MCF-7 and HCT116 cells. CK2 down-regulation-mediated SIRT1 inactivation promoted the degradation of inhibitors of NF-κB (IκB) by activating the AKT-IκB kinase (IKK) axis and increased the acetylation of lysine 310 on RelA/p65, an important site for the activity of NF-κB. kin-10 (the ortholog of CK2ß) knockdown increased zmp-1, -2, and -3 (the orthologs of MMP) expression in nematodes, but AKT inhibitor triciribine and SIRT activator resveratrol significantly abrogated the increased expression of these genes. Finally, antisense inhibitors of miR-186, miR-216b, miR-337-3p, and miR-760 suppressed CK2α down-regulation, activation of the AKT-IKK-NF-κB axis, RelA/p65 acetylation, and expression of SASP genes in cells treated with lipopolysaccharide. Therefore, this study indicated that CK2 down-regulation induces the expression of SASP factors through NF-κB activation, which is mediated by both activation of the SIRT1-AKT-IKK axis and RelA/p65 acetylation, suggesting that the mixture of the four miRNA inhibitors can be used as anti-inflammatory agents.


Assuntos
Regulação da Expressão Gênica/genética , Proteínas Serina-Treonina Quinases/metabolismo , Resveratrol/farmacologia , Ribonucleosídeos/farmacologia , Transdução de Sinais/genética , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Caseína Quinase II/genética , Caseína Quinase II/metabolismo , Linhagem Celular Tumoral , Senescência Celular , Regulação para Baixo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Metaloproteinase 3 da Matriz/metabolismo , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Sirtuína 1/metabolismo , Fator de Transcrição RelA/metabolismo , Quinase Induzida por NF-kappaB
3.
J Biochem Mol Toxicol ; 34(10): e22552, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32562591

RESUMO

A prior study identified that 4-O-methylascochlorin (MAC), a methylated derivative of ascochlorin (ASC) from the fungus Ascochyta viciae, activates autophagy in leukemia cells by suppressing c-Myc phosphorylation. However, the effects of MAC on autophagy in other cancer cells remain unknown. In the present study, we demonstrated that MAC activated autophagy in human glioblastoma. MAC increased expression of autophagy-related proteins, such as LC3-II and Beclin-1. Moreover, MAC stimulated AMP-activated protein kinase (AMPK) phosphorylation and suppressed phosphorylation of the mTOR, p70S6K, and 4EBP1. The well-known AMPK activator metformin increased LC3-II levels, which were augmented by MAC cotreatment. AMPK knockdown decreased LC3-II levels and inhibited MAC activation of autophagy. Furthermore, MAC suppression of c-Myc expression activated autophagy. Treatment with the c-MYC inhibitor, 10058-FA, induced autophagy, as did c-Myc small interfering RNA knockdown. These effects were augmented by MAC cotreatment. Taken together, these findings indicated that MAC induces autophagy in human glioblastoma by activating AMPK signaling and inhibiting c-Myc protein expression in human glioblastoma.


Assuntos
Adenilato Quinase/metabolismo , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Terpenos/farmacologia , Animais , Proteína Beclina-1/metabolismo , Neoplasias Encefálicas/enzimologia , Linhagem Celular Tumoral , Regulação para Baixo , Ativação Enzimática , Glioblastoma/enzimologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
4.
Biochem Biophys Res Commun ; 505(1): 67-73, 2018 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-30241941

RESUMO

Cellular senescence is an irreversible form of cell cycle arrest and senescent cells have a unique gene expression profile that is frequently accompanied by senescence-associated heterochromatic foci (SAHF). Here, we present evidence that CK2 downregulation induces trimethylation of histone H3 Lys 9 (H3K9me3), selective binding of HP1γ to H3K9me3, formation of SAHF, and reduction of cyclin D1 expression in HCT116 and MCF-7 cells. CK2 downregulation-mediated H3K9me3 is associated with induction of H3K9 trimethylase SUV39h1 as well as reduction of H3K9 dimethylase G9a and GLP in cells. In addition, Pharmacological inhibition of SUV39h1 and G9a overexpression significantly attenuated induction of senescence-associated ß-galactosidase (SA-ß-gal) activity, H3K9me3 and SAHF formation in CK2-downregulated cells. Moreover, CK2 downregulation induced H3K9me3 in nematodes. Taken together, these results demonstrate that CK2 downregulation leads to H3K9me3 and SAHF formation by increasing SUV39h1 and decreasing G9a.


Assuntos
Caseína Quinase II/metabolismo , Heterocromatina/metabolismo , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Caseína Quinase II/genética , Senescência Celular/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Heterocromatina/genética , Antígenos de Histocompatibilidade/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Humanos , Células MCF-7 , Metilação , Metiltransferases/genética , Interferência de RNA , Proteínas Repressoras/genética
5.
Biochem Biophys Res Commun ; 493(4): 1548-1554, 2017 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-28989024

RESUMO

We investigated the impact of protein kinase C (PKC) on cellular senescence. The PKC activity and expression of conventional PKC (cPKC) and atypical PKC (aPKC) isoforms decreased during replicative senescence in IMR-90 cells. Forced inhibition of cPKC or aPKC induced the activation of senescence markers, including senescence-associated ß-galactosidase activity and reactive oxygen species (ROS)-p53-p21Cip1/WAF1 axis in HCT116 and HEK293 cells. PKC inhibition triggered the nuclear exportation of FoxO3a via stimulation of AKT-mediated phosphorylation of FoxO3a, and thereby decreased the transcription of FoxO3a target genes. Conversely, ectopic expression of the PKC isoforms led to stimulation of the nuclear import of FoxO3a and expression of the FoxO3a target genes. Ectopic FoxO3a expression attenuated ROS accumulation and senescent phenotypes induced by PKC inhibition. Therefore, this study suggests for the first time that downregulation of PKC induces senescence through the AKT-FoxO3a-ROS-p53-p21Cip1/WAF1 pathway in HCT116 and HEK293 cells.


Assuntos
Senescência Celular/fisiologia , Proteína Forkhead Box O3/antagonistas & inibidores , Proteína Quinase C/metabolismo , Transporte Ativo do Núcleo Celular , Senescência Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação para Baixo , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Células HCT116 , Células HEK293 , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
6.
Proteomics ; 16(1): 136-49, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26449285

RESUMO

The zebrafish (Danio rerio) is a popular animal model used for studies on vertebrate development and organogenesis. Recent research has shown a similarity of approximately 70% between the human and zebrafish genomes and about 84% of human disease-causing genes have common ancestry with that of the zebrafish genes. Zebrafish embryos have a number of desirable features, including transparency, a large size, and rapid embryogenesis. Protein phosphorylation is a well-known PTM, which can carry out various biological functions. Recent MS developments have enabled the study of global phosphorylation patterns by using MS-based proteomics coupled with titanium dioxide phosphopeptide enrichment. In the present study, we identified 3500 nonredundant phosphorylation sites on 2166 phosphoproteins and quantified 1564 phosphoproteins in developing embryos of zebrafish. The distribution of Ser/Thr/Tyr phosphorylation sites in zebrafish embryos was found to be 88.9, 10.2, and 0.9%, respectively. A potential kinase motif was predicted using Motif-X analysis, for 80% of the identified phosphorylation sites, with the proline-directed motif appearing most frequently, and 35 phosphorylation sites having the pSF motif. In addition, we created six phosphoprotein clusters based on their dynamic pattern during the development of zebrafish embryos. Here, we report the largest dataset of phosphoproteins in zebrafish embryos and our results can be used for further studies on phosphorylation sites or phosphoprotein dynamics in zebrafish embryos.


Assuntos
Fosfoproteínas/análise , Proteoma/análise , Proteínas de Peixe-Zebra/análise , Peixe-Zebra/embriologia , Sequência de Aminoácidos , Animais , Cromatografia Líquida , Dados de Sequência Molecular , Fosfopeptídeos/análise , Fosforilação , Proteômica , Espectrometria de Massas em Tandem
7.
Apoptosis ; 21(5): 657-68, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26922069

RESUMO

4-O-Methyl-ascochlorin (MAC) is a methylated derivative of the prenyl-phenol antibiotic ascochlorin, which was isolated from an incomplete fungus, Ascochyta viciae. Although the effects of MAC on apoptosis have been reported, the underlying mechanisms remain unknown. Here, we show that MAC promoted apoptotic cell death and downregulated c-Myc expression in K562 human leukemia cells. The effect of MAC on apoptosis was similar to that of 10058-F4 (a c-Myc inhibitor) or c-Myc siRNA, suggesting that the downregulation of c-Myc expression plays a role in the apoptotic effect of MAC. Further investigation showed that MAC downregulated c-Myc by inhibiting protein synthesis. MAC promoted the phosphorylation of AMP-activated protein kinase (AMPK) and inhibited the phosphorylation of mammalian target of rapamycin (mTOR) and its target proteins, including p70S6 K and 4E-BP-1. Treatment of cells with AICAR (an AMPK activator), rapamycin (an mTOR inhibitor), or mTOR siRNA downregulated c-Myc expression and induced apoptosis to a similar extent to that of MAC. These results suggest that the effect of MAC on apoptosis induction in human leukemia cells is mediated by the suppression of c-Myc protein synthesis via an AMPK/mTOR-dependent mechanism.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antineoplásicos/farmacologia , Apoptose , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Terpenos/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Pontos de Checagem da Fase G1 do Ciclo Celular , Humanos , Células K562 , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/metabolismo , Serina-Treonina Quinases TOR/metabolismo
8.
Biochem Biophys Res Commun ; 478(1): 18-24, 2016 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-27470586

RESUMO

We previously showed that protein kinase CK2 downregulation mediates senescence through the reactive oxygen species (ROS)-p53-p21(Cip1/WAF1) pathway in various human cells. In the present study, we investigated whether the FoxO3a transcription factor is associated with ROS production during CK2 downregulation-induced senescence in human colon cancer HCT116 and breast cancer MCF-7 cells. FoxO3a overexpression suppressed ROS production and p53 stabilization induced by a CK2α knockdown. CK2α downregulation induced nuclear export of FoxO3a through stimulation of AKT-mediated phosphorylation of FoxO3a and decreased transcription of its target genes (Cu/ZnSOD, MnSOD, and catalase). In contrast, CK2α overexpression inhibited AKT-mediated FoxO3a phosphorylation. This resulted in nuclear accumulation of FoxO3a, and elevated expression of its target genes. Therefore, these data indicate for the first time that CK2 downregulation stimulates ROS generation by inhibiting FoxO3a during premature senescence in human colon and breast cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias do Colo/metabolismo , Proteína Forkhead Box O3/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transporte Ativo do Núcleo Celular , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Caseína Quinase II/genética , Caseína Quinase II/metabolismo , Senescência Celular , Colo/metabolismo , Colo/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Regulação para Baixo , Feminino , Proteína Forkhead Box O3/genética , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Células MCF-7 , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regulação para Cima
9.
Mol Ther ; 23(3): 445-55, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25502903

RESUMO

Recent evidence has shown that Ras homolog enriched in brain (Rheb) is dysregulated in Alzheimer's disease (AD) brains. However, it is still unclear whether Rheb activation contributes to the survival and protection of hippocampal neurons in the adult brain. To assess the effects of active Rheb in hippocampal neurons in vivo, we transfected neurons in the cornu ammonis 1 (CA1) region in normal adult rats with an adeno-associated virus containing the constitutively active human Rheb (hRheb(S16H)) and evaluated the effects on thrombin-induced neurotoxicity. Transduction with hRheb(S16H) significantly induced neurotrophic effects in hippocampal neurons through activation of mammalian target of rapamycin complex 1 (mTORC1) without side effects such as long-term potentiation impairment and seizures from the alteration of cytoarchitecture, and the expression of hRheb(S16H) prevented thrombin-induced neurodegeneration in vivo, an effect that was diminished by treatment with specific neutralizing antibodies against brain-derived neurotrophic factor (BDNF). In addition, our results showed that the basal mTORC1 activity might be insufficient to mediate the level of BDNF expression, but hRheb(S16H)-activated mTORC1 stimulated BDNF production in hippocampal neurons. These results suggest that viral vector transduction with hRheb(S16H) may have therapeutic value in the treatment of neurodegenerative diseases such as AD.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Região CA1 Hipocampal/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , Neurônios/metabolismo , Neuropeptídeos/genética , Transdução Genética/métodos , Animais , Anticorpos Neutralizantes/farmacologia , Fator Neurotrófico Derivado do Encéfalo/agonistas , Fator Neurotrófico Derivado do Encéfalo/antagonistas & inibidores , Fator Neurotrófico Derivado do Encéfalo/genética , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/efeitos dos fármacos , Dependovirus/genética , Dependovirus/metabolismo , Expressão Gênica , Vetores Genéticos/administração & dosagem , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/agonistas , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neuropeptídeos/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Ratos , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Trombina/antagonistas & inibidores , Trombina/toxicidade
10.
J Biol Chem ; 288(18): 12437-47, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23486471

RESUMO

The sensor histidine kinases of Mycobacterium tuberculosis, DosS and DosT, are responsible for sensing hypoxic conditions and consist of sensor and kinase cores responsible for accepting signals and phosphorylation activity, respectively. The kinase core contains a dimerization and histidine phosphate-accepting (DHp) domain and an ATP binding domain (ABD). The 13 histidine kinase genes of M. tuberculosis can be grouped based on the presence or absence of the ATP lid motif and F box (elements known to play roles in ATP binding) in their ABDs; DosS and DosT have ABDs lacking both these elements, and the crystal structures of their ABDs indicated that they were unsuitable for ATP binding, as a short loop covers the putative ATP binding site. Although the ABD alone cannot bind ATP, the kinase core is functional in autophosphorylation. Appropriate spatial arrangement of the ABD and DHp domain within the kinase core is required for both autophosphorylation and ATP binding. An ionic interaction between Arg(440) in the DHp domain and Glu(537) in the short loop of the ABD is available and may open the ATP binding site, by repositioning the short loop away from the site. Mutations at Arg(440) and Glu(537) reduce autophosphorylation activity. Unlike other histidine kinases containing an ATP lid, which protects bound ATP, DosS is unable to accept ATP until the ABD is properly positioned relative to the histidine; this may prevent unexpected ATP reactions. ATP binding can, therefore, function as a control mechanism for histidine kinase activity.


Assuntos
Trifosfato de Adenosina/química , Proteínas de Bactérias/química , Mycobacterium tuberculosis/enzimologia , Protamina Quinase/química , Trifosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Ativação Enzimática/fisiologia , Fosforilação/fisiologia , Protamina Quinase/metabolismo
11.
Biochem Biophys Res Commun ; 433(4): 420-5, 2013 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-23523798

RESUMO

Cellular senescence is a tumor suppression mechanism. We previously reported that CKII downregulation induces senescence in human lung fibroblast IMR-90 and colon cancer HCT116 cells. In this study, potential longevity drugs, including rapamycin, vitamin C, and vitamin E, blocked CKII downregulation-mediated senescence through reduction of reactive oxygen species (ROS) production in HCT116 cells. Since rapamycin is a mammalian target of rapamycin (mTOR) inhibitor, we examined the roles of mTOR and its upstream regulators phosphatidylinositol 3-kinase (PI3K) and AKT in CKII inhibition-mediated senescence. CKIIα knock-down or CKII inhibitor treatment strikingly increased phosphorylation of mTOR, p70S6K, an mTOR substrate, and AKT, whereas CKIIα overexpression reduced this phosphorylation event. This result indicated that CKII inhibition activated the PI3K-AKT-mTOR pathway. Further, pharmacological inhibition of PI3K and AKT attenuated ROS production and senescence in CKII-downregulated cells. Taken together, these results demonstrate, for the first time, that the PI3K-AKT-mTOR-ROS pathway is necessary for CKII inhibition-mediated cellular senescence.


Assuntos
Caseína Quinase II/metabolismo , Senescência Celular/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Ácido Ascórbico/farmacologia , Caseína Quinase II/antagonistas & inibidores , Ativação Enzimática , Ensaios Enzimáticos , Etídio/análogos & derivados , Etídio/metabolismo , Fluoresceínas/metabolismo , Fluorescência , Células HCT116 , Humanos , Peróxido de Hidrogênio/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Vitamina E/farmacologia
12.
Biochem Biophys Res Commun ; 429(3-4): 173-9, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23137536

RESUMO

We previously demonstrated that downregulation of protein kinase CKII induces cellular senescence in human colon cancer HCT116 cells. To investigate the role of microRNAs (miRNAs) in CKII downregulation during senescence, we employed computational algorithms. Four miRNAs (miR-186, miR-216b, miR-337-3p, and miR-760) were predicted to be miRNAs against CKIIα mRNA. Mimics of all four miRNAs jointly downregulated CKIIα expression in HCT116 cells. Reporter analysis and RT-PCR have suggested that these four miRNAs may stimulate degradation of CKIIα mRNA by targeting its 3' untranslated regions (UTRs). The four miRNA mimics increased senescent-associated ß-galactosidase (SA-ß-gal) staining, p53 and p21(Cip1/WAF1) expression, and reactive oxygen species (ROS) production. In contrast, concomitant knockdown of the four miRNAs by antisense inhibitors increased the CKIIα protein level and suppressed CKII inhibition-mediated senescence. Finally, CKIIα overexpression antagonized senescence induced by the four miRNA mimics. Therefore, the present results show that miR-186, miR-216b, miR-337-3p, and miR-760 cooperatively promote cellular senescence through the p53-p21(Cip1/WAF1) pathway by CKII downregulation-mediated ROS production in HCT116 cells.


Assuntos
Caseína Quinase II/biossíntese , Senescência Celular , Neoplasias Colorretais/enzimologia , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Caseína Quinase II/genética , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , MicroRNAs/genética , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/biossíntese , beta-Galactosidase/metabolismo
13.
BMB Rep ; 55(2): 92-97, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35000672

RESUMO

Lysine methylation is one of the most important histone modifications that modulate chromatin structure. In the present study, the roles of the histone lysine demethylases JMJD2a and LSD1 in CK2 downregulation-mediated senescence were investigated. The ectopic expression of JMJD2a and LSD1 suppressed the induction of senescence-associated ß-galactosidase activity and heterochromatin foci formation as well as the reduction of colony-forming and cell migration ability mediated by CK2 knockdown. CK2 downregulation inhibited JMJD2a and LSD1 expression by activating the mammalian target of rapamycin (mTOR)-ribosomal p70 S6 kinase (p70S6K) pathway. In addition, the downregulation of JMJD2a and LSD1 was involved in activating the p53-p21Cip1/WAF1-SUV39h1-trimethylation of the histone H3 Lys9 (H3K9me3) pathway in CK2-downregulated cells. Further, CK2 downregulation-mediated JMJD2a and LSD1 reduction was found to stimulate the dimethylation of Lys370 on p53 (p53K370me2) and nuclear import of SUV39h1. Therefore, this study indicated that CK2 downregulation reduces JMJD2a and LSD1 expression by activating mTOR, resulting in H3K9me3 induction by increasing the p53K370me2-dependent nuclear import of SUV39h1. These results suggest that CK2 is a potential therapeutic target for age-related diseases. [BMB Reports 2022;55(2): 92-97].


Assuntos
Senescência Celular , Proteína Supressora de Tumor p53 , Regulação para Baixo , Histona Desmetilases/metabolismo , Metilação , Proteína Supressora de Tumor p53/metabolismo
14.
Mol Cells ; 45(3): 112-121, 2022 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-34949740

RESUMO

Calorie restriction (CR) and the activation of autophagy extend healthspan by delaying the onset of age-associated diseases in most living organisms. Because protein kinase CK2 (CK2) downregulation induces cellular senescence and nematode aging, we investigated CK2's role in CR and autophagy. This study indicated that CR upregulated CK2's expression, thereby causing SIRT1 and AMP-activated protein kinase (AMPK) activation. CK2α overexpression, including antisense inhibitors of miR-186, miR-216b, miR-337-3p, and miR-760, stimulated autophagy initiation and nucleation markers (increase in ATG5, ATG7, LC3BII, beclin-1, and Ulk1, and decrease in SQSTM1/p62). The SIRT1 deacetylase, AKT, mammalian target of rapamycin (mTOR), AMPK, and forkhead homeobox type O (FoxO) 3a were involved in CK2-mediated autophagy. The treatment with the AKT inhibitor triciribine, the AMPK activator AICAR, or the SIRT1 activator resveratrol rescued a reduction in the expression of lgg-1 (the Caenorhabditis elegans ortholog of LC3B), bec-1 (the C. elegans ortholog of beclin-1), and unc-51 (the C. elegans ortholog of Ulk1), mediated by kin-10 (the C. elegans ortholog of CK2ß) knockdown in nematodes. Thus, this study indicated that CK2 acted as a positive regulator in CR and autophagy, thereby suggesting that these four miRs' antisense inhibitors can be used as CR mimetics or autophagy inducers.


Assuntos
Restrição Calórica , Caseína Quinase II , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Autofagia , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Caseína Quinase II/genética , Caseína Quinase II/metabolismo , Senescência Celular
15.
Oncogene ; 41(4): 550-559, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34785775

RESUMO

Cathepsin K is highly expressed in various types of cancers. However, the effect of cathepsin K inhibition in cancer cells is not well characterized. Here, cathepsin K inhibitor (odanacatib; ODN) and knockdown of cathepsin K (siRNA) enhanced oxaliplatin-induced apoptosis in multiple cancer cells through Bax upregulation. Bax knockdown significantly inhibited the combined ODN and oxaliplatin treatment-induced apoptotic cell death. Stabilization of p53 by ODN played a critical role in upregulating Bax expression at the transcriptional level. Casein kinase 2 (CK2)-dependent phosphorylation of OTUB1 at Ser16 played a critical role in ODN- and cathepsin K siRNA-mediated p53 stabilization. Interestingly, ODN-induced p53 and Bax upregulation were modulated by the production of mitochondrial reactive oxygen species (ROS). Mitochondrial ROS scavengers prevented OTUB1-mediated p53 stabilization and Bax upregulation by ODN. These in vitro results were confirmed by in mouse xenograft model, combined treatment with ODN and oxaliplatin significantly reduced tumor size and induced Bax upregulation. Furthermore, human renal clear carcinoma (RCC) tissues revealed a strong correlation between phosphorylation of OTUB1(Ser16) and p53/Bax expression. Our results demonstrate that cathepsin K inhibition enhances oxaliplatin-induced apoptosis by increasing OTUB1 phosphorylation via CK2 activation, thereby promoting p53 stabilization, and hence upregulating Bax.


Assuntos
Antineoplásicos/uso terapêutico , Catepsina K/metabolismo , Oxaliplatina/uso terapêutico , Proteína Supressora de Tumor p53/genética , Proteína X Associada a bcl-2/genética , Animais , Antineoplásicos/farmacologia , Apoptose , Morte Celular , Linhagem Celular Tumoral , Humanos , Camundongos , Oxaliplatina/farmacologia , Regulação para Cima
17.
BMB Rep ; 53(5): 272-277, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32317087

RESUMO

Protein kinase CK2 downregulation induces premature senescence in various human cell types via activation of the reactive oxygen species (ROS)-p53-p21Cip1/WAF1 pathway. The transcription factor "nuclear factor erythroid 2-related factor 2" (Nrf2) plays an important role in maintaining intracellular redox homeostasis. In this study, Nrf2 overexpression attenuated CK2 downregulation- induced ROS production and senescence markers including SA-ß-gal staining and activation of p53-p21Cip1/WAF1 in human breast (MCF-7) and colon (HCT116) cancer cells. CK2 downregulation reduced the transcription of Nrf2 target genes, such as glutathione S-transferase, glutathione peroxidase 2, and glutathione reductase 1. Furthermore, CK2 downregulation destabilized Nrf2 protein via inhibiting autophagic degradation of Kelch-like ECHassociated protein 1 (Keap1). Finally, CK2 downregulation decreased the nuclear import of Nrf2 by deactivating AMP-activated protein kinase (AMPK). Collectively, our data suggest that both Keap1 stabilization and AMPK inactivation are associated with decreased activity of Nrf2 in CK2 downregulation-induced cellular senescence. [BMB Reports 2020; 53(5): 272-277].


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia , Caseína Quinase II/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Humanos , Células Tumorais Cultivadas
18.
J Cell Biochem ; 107(2): 335-44, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19306296

RESUMO

Fibrosis in glomerulosclerosis causes progressive loss of renal function. Transforming growth factor (TGF)-beta, one of the major profibrotic cytokines, induces the synthesis of plasminogen activator inhibitor (PAI)-1, a factor that plays a crucial role in the development of fibrosis. Here, we found that an isoprenoid antibiotic, ascofuranone, suppresses expression of profibrotic factors including matrix proteins and PAI-1 induced by TGF-beta in renal fibroblasts. Ascofuranone selectively inhibits phosphorylation of epidermal growth factor receptor (EGFR), and downstream kinases such as Raf-1, MEK-1/2, and ERK-1/2. PAI-1 transcription also is suppressed by treatment with kinase inhibitors for MEK-1/2 or EGFR, and with small interfering RNA for EGFR. Ascofuranone inhibits cellular metalloproteinase activity, and an inhibitor of metalloproteinases suppresses EGFR phosphorylation and PAI-1 transcription. These results suggest that ascofuranone suppresses expression of profibrotic factors through the inhibition of an EGFR-dependent signal transduction pathway activated by metalloproteinases.


Assuntos
Antibacterianos/farmacologia , Receptores ErbB/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Sesquiterpenos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Western Blotting , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Rim/citologia , Rim/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Terpenos/farmacologia
19.
Mol Cells ; 42(11): 773-782, 2019 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-31617338

RESUMO

Cellular senescence is an irreversible form of cell cycle arrest. Senescent cells have a unique gene expression profile that is frequently accompanied by senescence-associated heterochromatic foci (SAHFs). Protein kinase CK2 (CK2) downregulation can induce trimethylation of histone H3 Lys 9 (H3K9me3) and SAHFs formation by activating SUV39h1. Here, we present evidence that the PI3K-AKTmTOR-reactive oxygen species-p53 pathway is necessary for CK2 downregulation-mediated H3K9me3 and SAHFs formation. CK2 downregulation promotes SUV39h1 stability by inhibiting its proteasomal degradation in a p53dependent manner. Moreover, the dephosphorylation status of Ser 392 on p53, a possible CK2 target site, enhances the nuclear import and subsequent stabilization of SUV39h1 by inhibiting the interactions between p53, MDM2, and SUV39h1. Furthermore, p21Cip1/WAF1 is required for CK2 downregulation-mediated H3K9me3, and dephosphorylation of Ser 392 on p53 is important for efficient transcription of p21Cip1/WAF1. Taken together, these results suggest that CK2 downregulation induces dephosphorylation of Ser 392 on p53, which subsequently increases the stability of SUV39h1 and the expression of p21Cip1/WAF1, leading to H3K9me3 and SAHFs formation.


Assuntos
Caseína Quinase II/metabolismo , Senescência Celular , Histonas/metabolismo , Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Caseína Quinase II/genética , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação para Baixo , Células HCT116 , Heterocromatina/genética , Humanos , Lisina/metabolismo , Células MCF-7 , Metilação , Fosforilação , Estabilidade Proteica , Interferência de RNA , Serina/metabolismo , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/genética
20.
BMB Rep ; 52(4): 265-270, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30103847

RESUMO

We investigated whether SIRT1 is associated with reactive oxygen species (ROS) accumulation during CK2 downregulationmediated senescence. SIRT1 overexpression suppressed ROS accumulation, reduced transcription of FoxO3a target genes, and nuclear export and acetylation of FoxO3a, which were induced by CK2 downregulation in HCT116 and MCF-7 cells. Conversely, overexpression of a dominant-negative mutant SIRT1 (H363Y) counteracted decreased ROS levels, increased transcriptional activity of FoxO3a, and increased nuclear import and decreased acetylation of FoxO3a, which were induced by CK2 upregulation. CK2 downregulation destabilized SIRT1 protein via an ubiquitin-proteasome pathway in human cells, whereas CK2 overexpression reduced ubiquitination of SIRT1. Finally, the SIRT1 activator resveratrol attenuated the accumulation of ROS and lipofuscin as well as lifespan shortening, and reduced expression of the DAF-16 target gene sod-3, which were induced by CK2 downregulation in nematodes. Altogether, this study demonstrates that inactivation of the SIRT1-FoxO3a axis, at least in part, is involved in ROS generation during CK2 downregulationmediated cellular senescence and nematode aging. [BMB Reports 2019; 52(4): 265-270].


Assuntos
Caenorhabditis elegans/metabolismo , Caseína Quinase II/metabolismo , Proteína Forkhead Box O3/metabolismo , Sirtuína 1/metabolismo , Envelhecimento/metabolismo , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Senescência Celular/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Células HCT116 , Humanos , Longevidade , Células MCF-7 , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Resveratrol/farmacologia , Transdução de Sinais , Sirtuína 1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA