Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 720: 150098, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-38749190

RESUMO

Telomerase reverse transcriptase (TERT) not only upholds telomeric equilibrium but also plays a pivotal role in multiple non-canonical cellular mechanisms, particularly in the context of aging, cancer, and genomic stability. Though depletion of SIRT1 in mouse embryonic fibroblasts has demonstrated telomere shortening, the impact of SIRT1 on enabling TERT to regulate telomeric homeostasis remains enigmatic. Here, we reveal that SIRT1 directly interacts with TERT, and promotes the nuclear localization and stability of TERT. Reverse transcriptase (RT) domain of TERT and N-terminus of SIRT1 mainly participated in their direct interaction. TERT, concomitantly expressed with intact SIRT1, exhibits nuclear localization, whereas TERT co-expressed with N-terminal-deleted SIRT1 remains in the cytosol. Furthermore, overexpression of SIRT1 enhances the nuclear localization and protein stability of TERT, akin to overexpression of deacetylase-inactive SIRT1, whereas N-terminal-deleted SIRT1 has no effect on TERT. These findings suggest a novel regulatory role of SIRT1 for TERT through direct interaction. This interaction provides new insights into the fields of aging, cancer, and genome stability governed by TERT and SIRT1.


Assuntos
Sirtuína 1 , Telomerase , Animais , Humanos , Camundongos , Núcleo Celular/metabolismo , Estabilidade Enzimática , Células HEK293 , Ligação Proteica , Estabilidade Proteica , Sirtuína 1/metabolismo , Sirtuína 1/genética , Telomerase/metabolismo , Telomerase/genética
2.
PLoS One ; 15(8): e0236881, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32745107

RESUMO

PIERCE1, p53 induced expression 1 in Rb null cells, is a novel p53 target involved in the DNA damage response and cell cycle in mice. These facts prompted us to study the function of PIERCE1 with respect to p53-associated pathophysiology of cancer in humans. Unexpectedly, PIERCE1 did not respond to overexpression and activation of p53 in humans. In this study, we swapped p53 protein expression in human and mouse cells to find the clue of this difference between species. Human p53 expression in mouse cells upregulated PIERCE1 expression, suggesting that p53-responsive elements on the PIERCE1 promoter are crucial, but not the p53 protein itself. Indeed, in silico analyses of PIERCE1 promoters revealed that p53-responsive elements identified in mice are not conserved in humans. Consistently, chromatin immunoprecipitation-sequencing (ChIP-seq) analyses confirmed p53 enrichment against the PIERCE1 promoter region in mice, not in human cells. To complement the p53 study in mice, further promoter analyses suggested that the human PIERCE1 promoter is more similar to guinea pigs, lemurs, and dogs than to rodents. Taken together, our results confirm the differential responsiveness of PIERCE1 expression to p53 due to species differences in PIERCE1 promoters. The results also show partial dissimilarity after p53 induction between mice and humans.


Assuntos
Proteínas de Ciclo Celular , Elementos de Resposta/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Dano ao DNA/genética , Humanos , Camundongos , Regiões Promotoras Genéticas , Transcrição Gênica/fisiologia
3.
Oncogene ; 39(36): 5876-5887, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32728173

RESUMO

KRAS-mutant non-small cell lung cancer (NSCLC) is a major lung cancer subtype that leads to many cancer-related deaths worldwide. Although numerous studies on KRAS-mutant type NSCLC have been conducted, new oncogenic or tumor suppressive genes need to be detected because a large proportion of NSCLC patients does not respond to currently used therapeutics. Here, we show the tumor-promoting function of a cell cycle-related protein, PIERCE1, in KRAS-mutant NSCLC. Mechanistically, PIERCE1 depletion inhibits cell growth and AKT phosphorylation (pAKT) at S473, which is particularly observed in KRAS-mutant lung cancers. Analyses of AKT-related genes using microarray, immunoblotting, and real-time quantitative PCR indicated that PIERCE1 negatively regulates the gene expression of the AKT suppressor, TRIB3, through the CHOP pathway, which is a key regulatory pathway for TRIB3 expression. Similarly, in vivo analyses of PIERCE1 depletion in the KRAS mutation-related lung cancer mouse models revealed the suppressive effect of PIERCE1 knockout in urethane- and KRASG12D-induced lung tumorigenesis with decreased pAKT levels observed in the tumors. Tissue microarrays of human lung cancers indicated the expression of PIERCE1 in 83% of lung cancers and its correlation with pAKT expression. Thus, we illustrate how PIERCE1 depletion may serve as a therapeutic strategy against KRAS-mutant NSCLC and propose the clinical benefit of PIERCE1.


Assuntos
Proteínas de Ciclo Celular/deficiência , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Mutação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais , Animais , Biomarcadores Tumorais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Prognóstico
4.
Exp Anim ; 69(4): 414-422, 2020 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-32581195

RESUMO

Colorectal cancer is the second most lethal cancer type across all ages and sexes, the many mechanisms of which are still currently being further elucidated. PIERCE1 has been known to be involved in the cell cycle and proliferation, the expression of which is regulated by stress conditions in a p53-dependent manner. Through a database search, we found that PIERCE1 was significantly augmented in patients with colorectal carcinoma compared to normal samples, suggesting its possible role in tumor regulation. Recently, PIERCE1 has also been reported to increase proliferation of a liver cancer cell line, indicating its possible role as an oncogene. To examine its relevance to tumorigenesis, such as whether it has either oncogenic or tumor suppressive function, PIERCE1 was knocked down and overexpressed in several colorectal cancer cell lines and mice, respectively. To evaluate the roles of Pierce1 in vivo, we established a Pierce1 transgenic (TG) mouse model and then administered azoxymethane with dextran sodium sulfate (DSS) to induce colorectal carcinogenesis via promoting mutations in Apc and Kras. Nonetheless, PIERCE1 depletion in these cell lines showed no significant change in cell growth. AOM/DSS-treated Pierce1 TG mice were comparable with respect to colon lengths, the number of polyps, and tumor sizes to those of the control mice. These results implicate that PIERCE1 does not play an oncogenic or tumor suppressive role in AOM/DSS-induced colorectal cancer.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorretais/genética , Expressão Gênica , Animais , Azoximetano/efeitos adversos , Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/patologia , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos Transgênicos , Proteína Supressora de Tumor p53/fisiologia
5.
Sci Rep ; 10(1): 4173, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-32144373

RESUMO

Engineered nucleases are widely used for creating frameshift or nonsense mutations in the target genes to eliminate gene functions. The resulting mRNAs carrying premature termination codons can be eliminated by nonsense-mediated mRNA decay. However, it is unclear how effective this process would be in vivo. Here, we found that the nonsense-mediated decay was unable to remove the mutant mRNAs in twelve out of sixteen homozygous mutant mice with frameshift mutations generated using engineered nucleases, which is far beyond what we expected. The frameshift mutant proteins translated by a single nucleotide deletion within the coding region were also detected in the p53 mutant mice. Furthermore, we showed that targeting the exons present downstream of the exons with a start codon or distant from ATG is relatively effective for eliminating mutant mRNAs in vivo, whereas the exons with a start codon are targeted to express the mutant mRNAs. Of the sixteen mutant mice generated, only four mutant mice targeting the downstream exons exhibited over 80% clearance of mutant mRNAs. Since the abnormal products, either mutant RNAs or mutant proteins, expressed by the target alleles might obscure the outcome of genome editing, these findings will provide insights in the improved performance of engineered nucleases when they are applied in vivo.


Assuntos
Edição de Genes , Fatores de Iniciação de Peptídeos/genética , RNA Mensageiro/genética , Animais , Western Blotting , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Feminino , Genótipo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Cell Death Dis ; 10(7): 511, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31263105

RESUMO

In ischemic human hearts, the induction of adenosine receptor A2B (ADORA2B) is associated with cardioprotection against ischemic heart damage, but the mechanism underlying this association remains unclear. Apaf-1-interacting protein (APIP) and ADORA2B transcript levels in human hearts are substantially higher in patients with heart failure than in controls. Interestingly, the APIP and ADORA2B mRNA levels are highly correlated with each other (R = 0.912). APIP expression was significantly increased in primary neonatal cardiomyocytes under hypoxic conditions and this induction reduced myocardial cell death via the activation of the AKT-HIF1α pathway. Accordingly, infarct sizes of APIP transgenic mice after left anterior descending artery ligation were significantly reduced compared to those of wild-type mice. Strikingly, knockdown of APIP expression impaired the cytoprotective effects of ADORA2B during hypoxic damage. Immunoprecipitation and proximity ligation assays revealed that APIP interacts with ADORA2B, leading to the stabilization of both proteins by interfering with lysosomal degradation, and to the activation of the downstream PKA-CREB signaling pathways. ADORA2B levels in the hearts of APIPTg/Tg, APIPTg/+, and Apip+/- mice were proportionally downregulated. In addition, ADORA2B D296G derived from the rs200741295 polymorphism failed to bind to APIP and did not exert cardioprotective activity during hypoxia. Moreover, Adora2b D296G knock-in mice were more vulnerable than control mice to myocardial infarction and intentional increases in APIP levels overcame the defective protection of the ADORA2B SNP against ischemic injury. Collectively, APIP is crucial for cardioprotection against myocardial infarction by virtue of binding to and stabilizing ADORA2B, thereby dampening ischemic heart injury.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptor A2B de Adenosina/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular , Células Cultivadas , Feminino , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Infarto do Miocárdio/genética , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Polimorfismo Genético/genética , Polimorfismo de Nucleotídeo Único/genética , Receptor A2B de Adenosina/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
7.
Biochem Biophys Res Commun ; 514(4): 1087-1092, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31097220

RESUMO

Etoposide-induced 2.4 kb transcript (EI24, also known as PIG8) is a p53 target gene involved in cell growth suppression and apoptosis and known to be frequently altered in human cancers. Although EI24 expression is decreased in various cancers and is associated with colorectal cancer progression and metastasis, the physiological function of EI24 in colorectal cancer is yet unclear. We generated an Ei24 conditional transgenic (Tg) mouse to study the therapeutic effects of Ei24 in vivo and evaluated whether Ei24 plays a role of a tumor suppressor using Ei24 Tg mouse crossed with ApcMin/+ mouse, which develops multiple intestinal adenomas. The overexpression of Ei24 failed to cause any notable difference in the number of polyps, lengths of the intestine and spleen, and survival rate between ApcMin/+ and ApcMin/+Ei24 Tg mice. Ei24 plays no significant role in colon cancer caused by the substitutional mutation of Apc in mice. Therefore, our result dismisses the hypothesized direct link between ApcMin/+ mutation and Ei24 expression in colorectal cancer model.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Proteínas Reguladoras de Apoptose/genética , Neoplasias Colorretais/genética , Modelos Animais de Doenças , Proteínas Nucleares/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Carcinogênese/genética , Neoplasias Colorretais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Proteínas Nucleares/metabolismo
8.
PLoS One ; 13(2): e0193182, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29462198

RESUMO

Autophagy is systematically regulated by upstream factors and nutrients. Recent studies reported that telomerase and hexokinase 2 [HK2) regulate autophagy through mTOR and that telomerase has the capacity to bind to the HK2 promoter. However, the molecular linkage among telomerase, HK2, and autophagy is not fully understood. Here, we show that HK2 connects telomerase to autophagy. HK2 inhibition in HepG2 cells suppressed TERT-induced autophagy activation and further enhancement by glucose deprivation. The HK2 downstream factor mTOR was responsible for the TERT-induced autophagy activation under glucose deprivation, implying that TERT promotes autophagy through an HK2-mTOR pathway. TERC played a role similar to that of TERT, and simultaneous expression of TERT and TERC synergistically enhanced HK2 expression and autophagy. At the gene level, TERT bound to the HK2 promoter at a specific region harboring the telomerase-responsive sequence 'TTGGG.' Mutagenesis of TERC and the TERT-responsive element in the HK2 promoter revealed that TERC is required for the binding of TERT to the HK2 promoter. We demonstrate the existence of a telomerase-HK2-mTOR-autophagy axis and suggest that inhibition of the interaction between telomerase and the HK2 promoter diminishes glucose starvation-induced autophagy.


Assuntos
Autofagia/fisiologia , Hexoquinase/metabolismo , Telomerase/metabolismo , Glucose/metabolismo , Glicólise/fisiologia , Células HeLa , Células Hep G2 , Hexoquinase/genética , Humanos , Regiões Promotoras Genéticas , Telomerase/genética
9.
Exp Anim ; 67(2): 229-237, 2018 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-29343656

RESUMO

Mouse models of obesity (ob/ob) and diabetes (db/db) in which the leptin (Lep) and leptin receptor (Lepr) genes have been mutated, respectively, have contributed to a better understanding of human obesity and type 2 diabetes and to the prevention, diagnosis, and treatment of these metabolic diseases. In this study, we report the first CRISPR-Cas9-induced Lep and Lepr knockout (KO) mouse models by co-microinjection of Cas9 mRNA and sgRNAs that specifically targeted Lep or Lepr in C57BL/6J embryos. Our newly established Lep and Lepr KO mouse models showed phenotypic disorders nearly identical to those found in ob/ob and db/db mice, such as an increase in body weight, hyperglycemia, and hepatic steatosis. Thus, Cas9-generated Lep and Lepr KO mouse lines will be easier for genotyping, to maintain the lines, and to use for future obesity and diabetes research.


Assuntos
Sistemas CRISPR-Cas/genética , Diabetes Mellitus Tipo 2 , Modelos Animais de Doenças , Obesidade , Animais , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/prevenção & controle , Diabetes Mellitus Tipo 2/terapia , Leptina/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microinjeções , Mutação , Obesidade/diagnóstico , Obesidade/genética , Obesidade/prevenção & controle , Obesidade/terapia , RNA Mensageiro/administração & dosagem , Receptores para Leptina/genética
10.
J Thorac Dis ; 10(Suppl 33): S3919-S3921, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30631516
11.
Lab Anim Res ; 34(4): 264-269, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30671114

RESUMO

Cell cycle dysfunction can cause severe diseases, including neurodegenerative disease and cancer. Mutations in cyclin-dependent kinase inhibitors controlling the G1 phase of the cell cycle are prevalent in various cancers. Mice lacking the tumor suppressors p16Ink4a (Cdkn2a, cyclin-dependent kinase inhibitor 2a), p19Arf (an alternative reading frame product of Cdkn2a,), and p27Kip1 (Cdkn1b, cyclin-dependent kinase inhibitor 1b) result in malignant progression of epithelial cancers, sarcomas, and melanomas, respectively. Here, we generated knockout mouse models for each of these three cyclin-dependent kinase inhibitors using engineered nucleases. The p16Ink4a and p19Arf knockout mice were generated via transcription activator-like effector nucleases (TALENs), and p27Kip1 knockout mice via clustered regularly interspaced short palindromic repeats/CRISPR-associated nuclease 9 (CRISPR/Cas9). These gene editing technologies were targeted to the first exon of each gene, to induce frameshifts producing premature termination codons. Unlike preexisting embryonic stem cell-based knockout mice, our mouse models are free from selectable markers or other external gene insertions, permitting more precise study of cell cycle-related diseases without confounding influences of foreign DNA.

12.
Lab Anim Res ; 34(4): 279-287, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30671116

RESUMO

Placenta specific 8 (PLAC8, also known as ONZIN) is a multi-functional protein that is highly expressed in the intestine, lung, spleen, and innate immune cells, and is involved in various diseases, including cancers, obesity, and innate immune deficiency. Here, we generated a Plac8 knockout mouse using the CRISPR/Cas9 system. The Cas9 mRNA and two single guide RNAs targeting a region near the translation start codon at Plac8 exon 2 were microinjected into mouse zygotes. This successfully eliminated the conventional translation start site, as confirmed by Sanger sequencing and PCR genotyping analysis. Unlike the previous Plac8 deficient models displaying increased adipose tissue and body weights, our male Plac8 knockout mice showed rather lower body weight than sex-matched littermate controls, though the only difference between these two mouse models is genetic context. Differently from the previously constructed embryonic stem cell-derived Plac8 knockout mouse that contains a neomycin resistance cassette, this knockout mouse model is free from a negative selection marker or other external insertions, which will be useful in future studies aimed at elucidating the multi-functional and physiological roles of PLAC8 in various diseases, without interference from exogenous foreign DNA.

13.
Autophagy ; 12(11): 2038-2053, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27541728

RESUMO

Historically, the ubiquitin-proteasome system (UPS) and autophagy pathways were believed to be independent; however, recent data indicate that these pathways engage in crosstalk. To date, the players mediating this crosstalk have been elusive. Here, we show experimentally that EI24 (EI24, autophagy associated transmembrane protein), a key component of basal macroautophagy/autophagy, degrades 14 physiologically important E3 ligases with a RING (really interesting new gene) domain, whereas 5 other ligases were not degraded. Based on the degradation results, we built a statistical model that predicts the RING E3 ligases targeted by EI24 using partial least squares discriminant analysis. Of 381 RING E3 ligases examined computationally, our model predicted 161 EI24 targets. Those targets are primarily involved in transcription, proteolysis, cellular bioenergetics, and apoptosis and regulated by TP53 and MTOR signaling. Collectively, our work demonstrates that EI24 is an essential player in UPS-autophagy crosstalk via degradation of RING E3 ligases. These results indicate a paradigm shift regarding the fate of E3 ligases.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Proteínas Nucleares/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Dano ao DNA , Metabolismo Energético , Humanos , Modelos Biológicos , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Domínios RING Finger , Reprodutibilidade dos Testes , Transdução de Sinais , Fator 2 Associado a Receptor de TNF/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína Supressora de Tumor p53/metabolismo
14.
Biochem Biophys Res Commun ; 478(3): 1198-204, 2016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27545609

RESUMO

Telomerase is a reverse transcriptase that consists of the telomerase RNA component (TERC) and the reverse transcriptase catalytic subunit (TERT) and specializes in the elongation of telomere ends. New evidence suggests that beyond classical telomere maintenance, TERT also possesses telomere length-independent functions that are executed via interaction with other binding proteins. One such reported TERT-interacting proteins is mTOR, a master nutrient sensor that is upregulated in several cancers; however, the physiological implications of the TERT-mTOR interaction in normal cellular processes as well as in tumorigenesis are poorly understood. Here, we report that TERT inhibits the kinase activity of mTOR complex 1 (mTORC1) in multiple cell lines, resulting in the activation of autophagy under both basal and amino acid-deprived conditions. Furthermore, TERT-deficient cells display the inability to properly execute the autophagy flux. Functionally, TERT-induced autophagy provides a survival advantage to cells in nutrient-deprived conditions. Collectively, these findings support a model in which gain of TERT function modulates mTORC1 activity and induces autophagy, which is required for metabolic rewiring to scavenge the nutrients necessary for fueling cancer cell growth in challenging tumor microenvironments.


Assuntos
Aminoácidos/deficiência , Autofagia , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Telomerase/metabolismo , Aminoácidos/metabolismo , Animais , Sobrevivência Celular , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Knockout
15.
Cell Rep ; 8(4): 991-8, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-25131203

RESUMO

Dilated cardiomyopathy (DCM) is a progressive heart disease characterized by left ventricular dilation and contractile dysfunction. Although many candidate genes have been identified with mouse models, few of them have been shown to be associated with DCM in humans. Germline depletion of Ncoa6, a nuclear hormone receptor coactivator, leads to embryonic lethality and heart defects. However, it is unclear whether Ncoa6 mutations cause heart diseases in adults. Here, we report that two independent mouse models of NCOA6 dysfunction develop severe DCM with impaired mitochondrial function and reduced activity of peroxisome proliferator-activated receptor δ (PPARδ), an NCOA6 target critical for normal heart function. Sequencing of NCOA6-coding regions revealed three independent nonsynonymous mutations present in 5 of 50 (10%) patients with idiopathic DCM (iDCM). These data suggest that malfunction of NCOA6 can cause DCM in humans.


Assuntos
Cardiomiopatia Dilatada/genética , Coativadores de Receptor Nuclear/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Células Cultivadas , Técnicas de Inativação de Genes , Humanos , Camundongos Transgênicos , Mitocôndrias Cardíacas/fisiologia , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Coativadores de Receptor Nuclear/metabolismo , PPAR delta/metabolismo , Transcrição Gênica
16.
Onco Targets Ther ; 6: 1161-6, 2013 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-24009427

RESUMO

Telomerase is responsible for maintaining the length of telomeres at the ends of chromosomes. Although most somatic cells do not exhibit telomerase activity, it is reactivated in approximately 85% of cancers. This simple and attractive phenomenon steers the development of anticancer drugs targeting telomeres and telomerase. Recent studies have been revealing extratelomeric roles of telomerase in normal tissues, affecting processes that are critical for survival and aging of organisms. In this review, we will discuss the current therapeutic strategies targeting telomeres and telomerase and evaluate their potential advantages and risks with respect to nontelomeric functions.

17.
J Biol Chem ; 288(43): 31261-7, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-24014029

RESUMO

The deficiency of retinoblastoma (Rb) gene deregulates E2F transcription factors and thus induces E2F target genes directly or p53 target genes indirectly via mouse p19(Arf) (or p14(ARF) in humans), an E2F target gene. Here, we identified that etoposide-induced 2.4 mRNA (Ei24)/p53-induced gene 8 (Pig8), a p53 target gene involved in apoptosis and autophagy, was up-regulated in Rb(-/-) mouse embryonic fibroblasts (MEFs). The Ei24 promoter was activated by E2F1 via multiple E2F-responsive elements, independently of the previously reported p53-responsive element. Chromatin immunoprecipitation assays revealed that E2F1 directly acts on the mouse Ei24 promoter. We observed that Ei24 expression was suppressed in p53(-/-) MEFs upon UVC irradiation, which was exacerbated in p53(-/-) E2f1(-/-) MEFs, supporting the positive role of E2F1 on Ei24 transcription. Furthermore, Ei24 knockdown sensitized p53(-/-) MEFs against UVC irradiation. Together, our data indicate that Ei24 is a novel E2F target gene contributing to the survival of p53-deficient cells upon UVC irradiation and thus may have a potential significance as a therapeutic target of certain chemotherapy for treating p53-deficient tumors.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Fator de Transcrição E2F1/metabolismo , Proteínas Nucleares/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta/efeitos adversos , Animais , Proteínas Reguladoras de Apoptose/genética , Morte Celular/genética , Morte Celular/efeitos da radiação , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Fator de Transcrição E2F1/genética , Humanos , Camundongos , Camundongos Knockout , Células NIH 3T3 , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Nucleares/genética , Elementos de Resposta/genética , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Transcrição Gênica/genética , Transcrição Gênica/efeitos da radiação , Proteína Supressora de Tumor p53/genética
18.
Int J Exp Pathol ; 92(4): 251-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21323770

RESUMO

p23 is a cochaperone of heat shock protein 90 and also interacts functionally with numerous steroid receptors and kinases. However, the in vivo roles of p23 remain unclear. To explore its in vivo function, we generated the transgenic (TG) mice ubiquitously overexpressing p23. The p23 TG mice spontaneously developed kidney abnormalities closely resembling human hydronephrosis. Consistently, kidney functions deteriorate significantly in the p23 TG mice compared to their wild-type (WT) littermates. Furthermore, the expression of target genes for aryl hydrocarbon receptor (AhR), such as cytochrome P450, family 1, subfamily A, polypeptide 1 (Cyp1A1) and cytochrome P450, family 1, subfamily B, polypeptide 1 (Cyp1B1), were induced in the kidneys of the p23 TG mice. These results indicate that the overexpression of p23 contributes to the development of hydronephrosis through the upregulation of the AhR pathway in vivo.


Assuntos
Regulação da Expressão Gênica/fisiologia , Hidronefrose/fisiopatologia , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/fisiologia , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1B1 , Modelos Animais de Doenças , Feminino , Hidronefrose/metabolismo , Rim/metabolismo , Rim/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Prostaglandina-E Sintases , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...