Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
J Am Soc Nephrol ; 32(8): 1987-2004, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34135081

RESUMO

BACKGROUND: Peripheral vascular diseases may induce chronic ischemia and cellular injury distal to the arterial obstruction. Cellular senescence involves proliferation arrest in response to stress, which can damage neighboring cells. Renal artery stenosis (RAS) induces stenotic-kidney dysfunction and injury, but whether these arise from cellular senescenceand their temporal pattern remain unknown. METHODS: Chronic renal ischemia was induced in transgenic INK-ATTAC and wild type C57BL/6 mice by unilateral RAS, and kidney function (in vivo micro-MRI) and tissue damage were assessed. Mouse healthy and stenotic kidneys were analyzed using unbiased single-cell RNA-sequencing. To demonstrate translational relevance, cellular senescence was studied in human stenotic kidneys. RESULTS: Using intraperitoneal AP20187 injections starting 1, 2, or 4 weeks after RAS, selective clearance of cells highly expressing p16Ink4a attenuated cellular senescence and improved stenotic-kidney function; however, starting treatment immediately after RAS induction was unsuccessful. Broader clearance of senescent cells, using the oral senolytic combination dasatinib and quercetin, in C57BL/6 RAS mice was more effective in clearing cells positive for p21 (Cdkn1a) and alleviating renal dysfunction and damage. Unbiased, single-cell RNA sequencing in freshly dissociated cells from healthy and stenotic mouse kidneys identified stenotic-kidney epithelial cells undergoing both mesenchymal transition and senescence. As in mice, injured human stenotic kidneys exhibited cellular senescence, suggesting this process is conserved. CONCLUSIONS: Maladaptive tubular cell senescence, involving upregulated p16 (Cdkn2a), p19 (Cdkn2d), and p21 (Cdkn1a) expression, is associated with renal dysfunction and injury in chronic ischemia. These findings support development of senolytic strategies to delay chronic ischemic renal injury.


Assuntos
Senescência Celular/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Isquemia/fisiopatologia , Rim/fisiopatologia , Insuficiência Renal Crônica/fisiopatologia , Quinases Ativadas por p21/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspase 8/metabolismo , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Doença Crônica , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Dasatinibe/farmacologia , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/fisiologia , Transição Epitelial-Mesenquimal , Expressão Gênica , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Humanos , Isquemia/etiologia , Rim/irrigação sanguínea , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteopontina/genética , Inibidores de Proteínas Quinases/farmacologia , Obstrução da Artéria Renal/complicações , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/patologia , Análise de Sequência de RNA , Análise de Célula Única , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia , Regulação para Cima , Quinases Ativadas por p21/genética
2.
Proc Natl Acad Sci U S A ; 115(13): 3344-3349, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29531090

RESUMO

Cell cycle progression is tightly regulated by cyclin-dependent kinases (CDKs). The ankyrin-repeat protein p19INK4d functions as a key regulator of G1/S transition; however, its molecular mode of action is unknown. Here, we combine cell and structural biology methods to unravel the mechanism by which p19INK4d controls cell cycle progression. We delineate how the stepwise phosphorylation of p19INK4d Ser66 and Ser76 by cell cycle-independent (p38) and -dependent protein kinases (CDK1), respectively, leads to local unfolding of the three N-terminal ankyrin repeats of p19INK4d This dissociates the CDK6-p19INK4d inhibitory complex and, thereby, activates CDK6. CDK6 triggers entry into S-phase, whereas p19INK4d is ubiquitinated and degraded. Our findings reveal how signaling-dependent p19INK4d unfolding contributes to the irreversibility of G1/S transition.


Assuntos
Ciclo Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p19/química , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Desdobramento de Proteína , Divisão Celular , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Fosforilação , Conformação Proteica , Proteólise , Transdução de Sinais
3.
Blood Cells Mol Dis ; 85: 102477, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32711219

RESUMO

Chronic myeloid leukemia (CML) is a kind of myeloproliferative disorder caused by a constitutively active BCR-ABL tyrosine kinase. Tyrosine kinase inhibitors (TKIs), imatinib and its derivatives, have achieved great progress in the treatment of CML. However, many CML patients do not respond to TKIs alone. p19INK4d, a cyclin-dependent kinase inhibitor, plays important roles in proliferation, DNA damage repair, apoptosis and cell differentiation, but its role in CML is unknown. Herein, we found that the expression of p19INK4d in CML patients was significantly lower than that in healthy controls. p19INK4d overexpression inhibits cell proliferation through cell cycle arrest, and cooperates with imatinib to inhibit CML more effectively in vitro and in vivo. Mechanistically, p19INK4d decreased the expression of BCR-ABL and its downstream molecules p-Mek1/2, moreover, the expression of Gli-1, c-myc, MUC1, Shh and TC48 also reduced significantly. Collectively, p19INK4d inhibits proliferation and enhances imatinib efficacy in the treatment of CML. These findings maybe have implications for developing potential targets to increase imatinib sensitivity for CML.


Assuntos
Antineoplásicos/uso terapêutico , Inibidor de Quinase Dependente de Ciclina p19/genética , Proteínas de Fusão bcr-abl/genética , Regulação Leucêmica da Expressão Gênica , Mesilato de Imatinib/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Proteínas de Fusão bcr-abl/metabolismo , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Mesilato de Imatinib/farmacologia , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos
4.
Semin Cell Dev Biol ; 65: 96-105, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27864084

RESUMO

Vestibular sensation is essential for gaze stabilization, balance, and perception of gravity. The vestibular receptors in mammals, Type I and Type II hair cells, are located in five small organs in the inner ear. Damage to hair cells and their innervating neurons can cause crippling symptoms such as vertigo, visual field oscillation, and imbalance. In adult rodents, some Type II hair cells are regenerated and become re-innervated after damage, presenting opportunities for restoring vestibular function after hair cell damage. This article reviews features of vestibular sensory cells in mammals, including their basic properties, how they develop, and how they are replaced after damage. We discuss molecules that control vestibular hair cell regeneration and highlight areas in which our understanding of development and regeneration needs to be deepened.


Assuntos
Linhagem da Célula/genética , Sensação Gravitacional/fisiologia , Células Ciliadas Vestibulares/citologia , Equilíbrio Postural/fisiologia , Regeneração/genética , Animais , Diferenciação Celular , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p19/genética , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Ciliadas Auditivas/citologia , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Vestibulares/classificação , Células Ciliadas Vestibulares/metabolismo , Camundongos , Organogênese/genética , Transdução de Sinais , beta Catenina/genética , beta Catenina/metabolismo
5.
Blood ; 129(2): 226-237, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-27879259

RESUMO

Terminal erythroid differentiation is tightly coordinated with cell-cycle exit, which is regulated by cyclins, cyclin-dependent kinases, and cyclin-dependent kinase inhibitors (CDKI), yet their roles in erythropoiesis remain to be fully defined. We show here that p19INK4d, a member of CDKI family, is abundantly expressed in erythroblasts and that p19INK4d knockdown delayed erythroid differentiation, inhibited cell growth, and led to increased apoptosis and generation of abnormally nucleated late-stage erythroblasts. Unexpectedly, p19INK4d knockdown did not affect cell cycle. Rather, it led to decreased expression of GATA1 protein. Importantly, the differentiation and nuclear defects were rescued by ectopic expression of GATA1. Because the GATA1 protein is protected by nuclear heat shock protein family (HSP) member HSP70, we examined the effects of p19INK4d knockdown on HSP70 and found that p19INK4d knockdown led to decreased expression of HSP70 and its nuclear localization. The reduced levels of HSP70 are the result of reduced extracellular signal-regulated kinase (ERK) activation. Further biochemical analysis revealed that p19INK4d directly binds to Raf kinase inhibitor PEBP1 and that p19INK4d knockdown increased the expression of PEBP1, which in turn led to reduced ERK activation. Thus we have identified an unexpected role for p19INK4d via a novel PEBP1-p-ERK-HSP70-GATA1 pathway. These findings are likely to have implications for improved understanding of disordered erythropoiesis.


Assuntos
Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Eritropoese/fisiologia , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica/fisiologia , Western Blotting , Células Cultivadas , Sangue Fetal , Citometria de Fluxo , Imunofluorescência , Técnicas de Silenciamento de Genes , Humanos , Imunoprecipitação , Reação em Cadeia da Polimerase , Transdução de Sinais/fisiologia
6.
Development ; 142(1): 41-50, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25480918

RESUMO

Genetic data indicate that abrogation of Notch-Rbpj or Wnt-ß-catenin pathways results in the loss of the intestinal stem cells (ISCs). However, whether the effect of Notch is direct or due to the aberrant differentiation of the transit-amplifying cells into post-mitotic goblet cells is unknown. To address this issue, we have generated composite tamoxifen-inducible intestine-specific genetic mouse models and analyzed the expression of intestinal differentiation markers. Importantly, we found that activation of ß-catenin partially rescues the differentiation phenotype of Rbpj deletion mutants, but not the loss of the ISC compartment. Moreover, we identified Bmi1, which is expressed in the ISC and progenitor compartments, as a gene that is co-regulated by Notch and ß-catenin. Loss of Bmi1 resulted in reduced proliferation in the ISC compartment accompanied by p16(INK4a) and p19(ARF) (splice variants of Cdkn2a) accumulation, and increased differentiation to the post-mitotic goblet cell lineage that partially mimics Notch loss-of-function defects. Finally, we provide evidence that Bmi1 contributes to ISC self-renewal.


Assuntos
Intestinos/patologia , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Compartimento Celular , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p19/genética , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Reparo do DNA , Homeostase , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/deficiência , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Intestinos/anormalidades , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Complexo Repressor Polycomb 1/deficiência , Complexo Repressor Polycomb 1/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Receptores Notch/deficiência , Ativação Transcricional/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
7.
Hum Mol Genet ; 23(25): 6826-37, 2014 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-25104850

RESUMO

Uncontrolled cell cycle entry, resulting from deregulated CDK-RB1-E2F pathway activity, is a crucial determinant of neuroblastoma cell malignancy. Here we identify neuroblastoma-suppressive functions of the p19-INK4d CDK inhibitor and uncover mechanisms of its repression in high-risk neuroblastomas. Reduced p19-INK4d expression was associated with poor event-free and overall survival and neuroblastoma risk factors including amplified MYCN in a set of 478 primary neuroblastomas. High MYCN expression repressed p19-INK4d mRNA and protein levels in different neuroblastoma cell models with conditional MYCN expression. MassARRAY and 450K methylation analyses of 105 primary neuroblastomas uncovered a differentially methylated region within p19-INK4d. Hypermethylation of this region was associated with reduced p19-INK4d expression. In accordance, p19-INK4d expression was activated upon treatment with the demethylating agent, 2'-deoxy-5-azacytidine, in neuroblastoma cell lines. Ectopic p19-INK4d expression decreased viability, clonogenicity and the capacity for anchorage-independent growth of neuroblastoma cells, and shifted the cell cycle towards the G1/0 phase. p19-INK4d also induced neurite-like processes and markers of neuronal differentiation. Moreover, neuroblastoma cell differentiation, induced by all-trans retinoic acid or NGF-NTRK1-signaling, activated p19-INK4d expression. Our findings pinpoint p19-INK4d as a neuroblastoma suppressor and provide evidence for MYCN-mediated repression and for epigenetic silencing of p19-INK4d by DNA hypermethylation in high-risk neuroblastomas.


Assuntos
Inibidor de Quinase Dependente de Ciclina p19/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias do Sistema Nervoso/genética , Neuroblastoma/genética , Neurônios/metabolismo , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Adolescente , Adulto , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Criança , Pré-Escolar , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Metilação de DNA/efeitos dos fármacos , Decitabina , Epigênese Genética , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Proteína Proto-Oncogênica N-Myc , Estadiamento de Neoplasias , Neoplasias do Sistema Nervoso/metabolismo , Neoplasias do Sistema Nervoso/mortalidade , Neoplasias do Sistema Nervoso/patologia , Neuroblastoma/metabolismo , Neuroblastoma/mortalidade , Neuroblastoma/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Transdução de Sinais , Análise de Sobrevida , Tretinoína/farmacologia
8.
Biochim Biophys Acta ; 1840(7): 2171-83, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24667034

RESUMO

BACKGROUND: During evolution, organisms with renewable tissues have developed mechanisms to prevent tumorigenesis, including cellular senescence and apoptosis. Cellular senescence is characterized by a permanent cell cycle arrest triggered by both endogenous stress and exogenous stress. The p19INK4d, a member of the family of cyclin-dependent kinase inhibitors (INK4), plays an important role on cell cycle regulation and in the cellular DNA damage response. We hypothesize that p19INK4d is a potential factor involved in the onset and/or maintenance of the senescent state. METHODS: Senescence was confirmed by measuring the cell cycle arrest and the senescence-associated ß-galactosidase activity. Changes in p19INK4d expression and localization during senescence were determined by Western blot and immunofluorescence assays. Chromatin condensation was measured by microccocal nuclease digestion and histone salt extraction. RESULTS: The data presented here show for the first time that p19INK4d expression is up-regulated by different types of senescence. Changes in senescence-associated hallmarks were driven by modulation of p19 expression indicating a direct link between p19INK4d induction and the establishment of cellular senescence. Following a senescence stimulus, p19INK4d translocates to the nucleus and tightly associates with chromatin. Moreover, reduced levels of p19INK4d impair senescence-related global genomic heterochromatinization. Analysis of p19INK4d mRNA and protein levels in tissues from differently aged mice revealed an up-regulation of p19INK4d that correlates with age. CONCLUSION: We propose that p19INK4d participates in the cellular mechanisms that trigger senescence by contributing to chromatin compaction. GENERAL SIGNIFICANCE: This study provides novel insights into the dynamics process of cellular senescence, a central tumor suppressive mechanism.


Assuntos
Envelhecimento/genética , Senescência Celular/genética , Inibidor de Quinase Dependente de Ciclina p19/genética , Heterocromatina/genética , Animais , Carcinogênese , Pontos de Checagem do Ciclo Celular/genética , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Dano ao DNA/genética , Regulação da Expressão Gênica , Camundongos , beta-Galactosidase/biossíntese
9.
Biochim Biophys Acta ; 1843(7): 1309-24, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24703879

RESUMO

DNA damage, which perturbs genomic stability, has been linked to cognitive decline in the aging human brain, and mutations in DNA repair genes have neurological implications. Several studies have suggested that DNA damage is also increased in brain disorders such as Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. However, the precise mechanisms connecting DNA damage with neurodegeneration remain poorly understood. CDK5, a critical enzyme in the development of the central nervous system, phosphorylates a number of synaptic proteins and regulates dendritic spine morphogenesis, synaptic plasticity and learning. In addition to these physiological roles, CDK5 has been involved in the neuronal death initiated by DNA damage. We hypothesized that p19INK4d, a member of the cell cycle inhibitor family INK4, is involved in a neuroprotective mechanism activated in response to DNA damage. We found that in response to genotoxic injury or increased levels of intracellular calcium, p19INK4d is transcriptionally induced and phosphorylated by CDK5 which provides it with greater stability in postmitotic neurons. p19INK4d expression improves DNA repair, decreases apoptosis and increases neuronal survival under conditions of genotoxic stress. Our in vivo experiments showed that decreased levels of p19INK4d rendered hippocampal neurons more sensitive to genotoxic insult resulting in the loss of cognitive abilities that rely on the integrity of this brain structure. We propose a feedback mechanism by which the neurotoxic effects of CDK5-p25 activated by genotoxic stress or abnormal intracellular calcium levels are counteracted by the induction and stabilization of p19INK4d protein reducing the adverse consequences on brain functions.


Assuntos
Cálcio/metabolismo , Quinase 5 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Reparo do DNA/genética , Hipocampo/metabolismo , Neurônios/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Cognição/fisiologia , Quinase 5 Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p19/genética , Citotoxinas/farmacologia , Dano ao DNA , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Humanos , Camundongos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Transdução de Sinais , Transcrição Gênica , Zinostatina/farmacologia
10.
Mol Cell Biochem ; 398(1-2): 63-72, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25204969

RESUMO

DNA is continuously exposed to damaging agents that can lead to changes in the genetic information with adverse consequences. Nonetheless, eukaryotic cells have mechanisms such as the DNA damage response (DDR) to prevent genomic instability. The DNA of eukaryotic cells is packaged into nucleosomes, which fold the genome into highly condensed chromatin, but relatively little is known about the role of chromatin accessibility in DNA repair. p19INK4d, a cyclin-dependent kinase inhibitor, plays an important role in cell cycle regulation and cellular DDR. Extensive data indicate that p19INK4d is a critical factor in the maintenance of genomic integrity and cell survival. p19INK4d is upregulated by various genotoxics, improving the repair efficiency for a variety of DNA lesions. The evidence of p19INK4d translocation into the nucleus and its low sequence specificity in its interaction with DNA prompted us to hypothesize that p19INK4d plays a role at an early stage of cellular DDR. In the present study, we demonstrate that upon oxidative DNA damage, p19INK4d strongly binds to and relaxes chromatin. Furthermore, in vitro accessibility assays show that DNA is more accessible to a restriction enzyme when a chromatinized plasmid is incubated in the presence of a protein extract with high levels of p19INK4d. Nuclear protein extracts from cells overexpressing p19INK4d are better able to repair a chromatinized and damaged plasmid. These observations support the notion that p19INK4d would act as a chromatin accessibility factor that allows the access of the repair machinery to the DNA damage site.


Assuntos
Cromatina/metabolismo , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Dano ao DNA , Estresse Oxidativo , Transporte Ativo do Núcleo Celular , Animais , Northern Blotting , Western Blotting , Linhagem Celular , Núcleo Celular/metabolismo , Cromatina/genética , Inibidor de Quinase Dependente de Ciclina p19/genética , Reparo do DNA , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Células HeLa , Humanos , Microscopia Confocal , Ligação Proteica
11.
J Pathol ; 233(2): 113-23, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24648314

RESUMO

Cullin4A (Cul4A) is a scaffold protein that assembles cullin-RING ubiquitin ligase (E3) complexes and regulates many cellular events, including cell survival, development, growth and cell cycle control. Our previous study suggested that Cul4A is oncogenic in vitro, but its oncogenic role in vivo has not been studied. Here, we used a Cul4A transgenic mouse model to study the potential oncogenic role of Cul4A in lung tumour development. After Cul4A over-expression was induced in the lungs for 32 weeks, atypical epithelial cells were observed. After 40 weeks, lung tumours were visible and were characterized as grade I or II adenocarcinomas. Immunohistochemistry (IHC) revealed decreased levels of Cul4A-associated proteins p21(CIP1) and tumour suppressor p19(ARF) in the lung tumours, suggesting that Cul4A regulated their expression in these tumours. Increased levels of p27(KIP1) and p16(INK4a) were also detected in these tumours. Moreover, the protein level of DNA replication licensing factor CDT1 was decreased. Genomic instability in the lung tumours was further analysed by the results from pericentrin protein expression and array comparative genomic hybridization analysis. Furthermore, knocking down Cul4A expression in lung cancer H2170 cells increased their sensitivity to the chemotherapy drug cisplatin in vitro, suggesting that Cul4A over-expression is associated with cisplatin resistance in the cancer cells. Our findings indicate that Cul4A is oncogenic in vivo, and this Cul4A mouse model is a tool in understanding the mechanisms of Cul4A in human cancers and for testing experimental therapies targeting Cul4A.


Assuntos
Adenocarcinoma/metabolismo , Transformação Celular Neoplásica/metabolismo , Proteínas Culina/metabolismo , Neoplasias Pulmonares/metabolismo , Pulmão/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Cisplatino/farmacologia , Proteínas Culina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Instabilidade Genômica , Humanos , Pulmão/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Transgênicos , Gradação de Tumores , Antígeno Nuclear de Célula em Proliferação/metabolismo , Interferência de RNA , Fatores de Tempo , Transfecção , Regulação para Cima
12.
Mol Vis ; 20: 215-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24623965

RESUMO

PURPOSE: Persistent hyperplastic primary vitreous (PHPV) represents a developmental eye disease known to have diverse manifestations ranging from a trivial remnant of hyaloid vessels to a dense fibrovascular mass causing lens opacity and retinal detachment. PHPV can be modeled in mice lacking individual genes, but certain features of such models differ from the clinical realm. For example, mice lacking the Arf gene have uniformly severe disease with consistent autosomal recessive disease penetrance. We tested whether the graded somatic loss of Arf in a subset of cells in chimeric mice mimics the range of disease in a non-heritable manner. METHODS: Wild type ↔ Arf(-/-) mouse chimeras were generated by morulae fusion, and when the mice were 10 weeks old, fundoscopic, slit-lamp, and histological evaluations were performed. The relative fraction of cells of the Arf(-/-) lineage was assessed with visual, molecular genetic, and histological analysis. Objective quantification of various aspects of the phenotype was correlated with the genotype. RESULTS: Sixteen chimeras were generated and shown to have low, medium, and high contributions of Arf(-/-) cells to tail DNA, the cornea, and the retinal pigment epithelium (RPE), with excellent correlation between chimerism in the tail DNA and the RPE. Phenotypic differences (coat color and severity of eye disease) were evident, objectively quantified, and found to correlate with the contribution of Arf(-/-) cells to the RPE and tail-derived DNA, but not the cornea. CONCLUSIONS: Generating animals composed of different numbers of Arf(-/-) cells mimicked the range of disease severity observed in patients with PHPV. This establishes the potential for full manifestations of PHPV to be caused by somatic mutations of a single gene during development.


Assuntos
Inibidor de Quinase Dependente de Ciclina p19/genética , Deleção de Genes , Mosaicismo , Vítreo Primário Hiperplásico Persistente/genética , Vítreo Primário Hiperplásico Persistente/patologia , Animais , Linhagem da Célula , Quimera , Inibidor de Quinase Dependente de Ciclina p19/deficiência , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , DNA/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Oftalmoscopia , Fenótipo , Pigmentação , Epitélio Pigmentado da Retina/patologia
13.
Clin Exp Pharmacol Physiol ; 41(12): 965-75, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25345622

RESUMO

Cellular senescence protects multicellular organisms from tissue overgrowth including cancer, and contributes to tissue ageing. With stable cell cycle arrests, cellular senescence has been mostly studied in the adult tissues of mammals. In the present study, we report widespread cellular senescence within certain time windows of late-phase normal development of mouse embryos. Using in situ senescence-associated ß-galactosidase (SA-ß-gal) staining, we showed SA-ß-gal activity in selected cell populations of the brain, stomach, interdigital webs, tail, ear, limbs and nasal mouth area on gestation day 14.5 of the mouse embryos. On day 18.5 of gestation, selected cells in the intestines and bone developmental areas showed SA-ß-gal activity. The chondrocytes in ossification zones were significantly marked by the activities of SA-ß-gal, p21, p15 and Hp1Y, suggesting activation of the cell cycle checkpoint by the p53 and Rb pathways, and development of senescence-associated heterochromatic foci. Throughout gestation days 14.5-18.5, the trophoblast cells in the labyrinth layer of the placentas also showed strong activities of SA-ß-gal, p53 and p21. Increased expressions of p19, p16 and Rb of the p16/Rb pathway, and reduced expressions of Ki67 were also observed in the placentas. Taken together, the present findings suggest that cellular senescence represents an essential mechanism at multiple sites including the fetal bone forming zones and placenta during mammalian embryonic development, playing potential roles in the full embryonic development of tissue growth and organ formation.


Assuntos
Senescência Celular/fisiologia , Desenvolvimento Embrionário/fisiologia , Feto/fisiologia , Animais , Pontos de Checagem do Ciclo Celular/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Feminino , Feto/metabolismo , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína Supressora de Tumor p53/metabolismo , beta-Galactosidase/metabolismo , Quinases Ativadas por p21/metabolismo
14.
Genet Mol Res ; 13(4): 10086-96, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25501220

RESUMO

The elucidation of the molecular mechanisms underlying the effects of traditional Chinese medicines in clinical practice is a key step toward their worldwide application, and this topic is currently a subject of intense research interest. Rg1, a component of ginsenoside, has recently been shown to perform several pharmacological functions; however, the underlying mechanisms of these effects remain unclear. In the present study, we investigated whether Rg1 has an anti-senescence effect on hematopoietic stem cells (HSCs) and the possible molecular mechanisms driving any effects. The results showed that Rg1 could effectively delay tert-butyl hydroperoxide (t-BHP)-induced senescence and inhibit gene expression in the p16(INK4a)-Rb and p19(Arf)-p53-p21(Cip/Waf1) signaling pathways in HSCs. Our study suggested that these two signaling pathways might be potential targets for elucidating the molecular mechanisms of the Rg1 anti-senescence effect.


Assuntos
Senescência Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p19/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Ginsenosídeos/farmacologia , Células-Tronco Hematopoéticas/citologia , Proteína do Retinoblastoma/genética , Proteína Supressora de Tumor p53/genética , Animais , Senescência Celular/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Ginsenosídeos/química , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/metabolismo , beta-Galactosidase/metabolismo , terc-Butil Hidroperóxido/farmacologia
15.
Biochem Biophys Res Commun ; 441(1): 196-201, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24140061

RESUMO

Nucleostemin is a nucleolar protein known to play a variety of roles in cell-cycle progression, apoptosis inhibition, and DNA damage protection in embryonic stem cells and tissue stem cells. However, the role of nucleostemin in hematopoietic stem cells (HSCs) is yet to be determined. Here, we identified an indispensable role of nucleostemin in mouse HSCs. Depletion of nucleostemin using short hairpin RNA strikingly impaired the self-renewal activity of HSCs both in vitro and in vivo. Consistently, nucleostemin depletion triggered apoptosis rather than cell-cycle arrest in HSCs. Furthermore, DNA damage accumulated during cultivation upon depletion of nucleostemin. The impaired self-renewal activity of HSCs induced by nucleostemin depletion was partially rescued by p53 deficiency but not by p16(Ink4a) or p19(Arf) deficiency. Taken together, our study demonstrates that nucleostemin protects HSCs from DNA damage accumulation and is required for the maintenance of HSCs.


Assuntos
Proteínas de Transporte/metabolismo , Instabilidade Genômica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Nucleares/metabolismo , Animais , Apoptose , Células da Medula Óssea/metabolismo , Ciclo Celular , Ensaio de Unidades Formadoras de Colônias , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Dano ao DNA , Proteínas de Ligação ao GTP , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA , Proteína Supressora de Tumor p53/metabolismo
16.
Genes Cells ; 17(8): 673-87, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22747700

RESUMO

Human amniotic fluid-derived mesenchymal stromal cells (hAMSC) have become one of the main cell populations used in regenerative medicine and for the study of various clinical disorders. These cells have a great capacity for proliferation and differentiation and do not form teratomas when transplanted into animal models, and their stemness seems to be between embryonic cells and adult mesenchymal cells. Before their use in cell therapy, they must be cultured and expanded in vitro, but the effect this process has on their fitness, a determining factor for the success or failure of cell therapy, is unknown. We undertook a follow-up of gene and microRNAs (miRNAs) expression using microarray of hAMSC for the first 15 passages. Significant changes were noted in the expression of various mRNAs and miRNAs, particularly down-regulation of TP53, increased expression of hsa-miR-125a and up-regulation of CDKN2D . The variations in TP53 and hsa-miR-125a may act as an indicator of the stemness of the hAMSC, whereas CDKN2D may indicate the begging of early senescence process in a p53-independent mechanism. The genes described in this study will help evaluate the fitness of hAMSC, thus guaranteeing their biological quality for use in regenerative medicine.


Assuntos
Líquido Amniótico/citologia , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Líquido Amniótico/metabolismo , Adesão Celular , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Biologia Computacional/métodos , Inibidor de Quinase Dependente de Ciclina p19/genética , Regulação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Medicina Regenerativa/métodos , Proteína Supressora de Tumor p53/genética
17.
Birth Defects Res A Clin Mol Teratol ; 97(2): 69-78, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23281115

RESUMO

Congenital human cytomegalovirus (CMV) infection is the leading nongenetic etiology of sensorineural hearing loss (SNHL) at birth and prelingual SNHL not expressed at birth. The paucity of temporal bone autopsy specimens from infants with congenital CMV infection has hindered the critical correlation of histopathology with pathogenesis. Here, we present an in vitro embryonic mouse model of CMV-infected cochleas that mimics the human sites of viral infection and associated pathology. There is a striking dysplasia/hyperplasia in mouse CMV-infected cochlear epithelium and mesenchyme, including organ of Corti hair and supporting cells and stria vascularis. This is concomitant with significant dysregulation of p19, p21, p27, and Pcna gene expression, as well as proliferating cell nuclear antigen (PCNA) protein expression. Other pathologies similar to those arising from known deafness gene mutations include downregulation of KCNQ1 protein expression in the stria vascularis, as well as hypoplastic and dysmorphic melanocytes. Thus, this model provides a relevant and reliable platform within which the detailed cell and molecular biology of CMV-induced deafness may be studied.


Assuntos
Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/patologia , Camundongos , Órgão Espiral/anormalidades , Estria Vascular/patologia , Animais , Inibidor de Quinase Dependente de Ciclina p19/genética , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Modelos Animais de Doenças , Feto , Regulação da Expressão Gênica no Desenvolvimento , Perda Auditiva Neurossensorial/congênito , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Humanos , Lactente , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Melanócitos/metabolismo , Melanócitos/patologia , Muromegalovirus/fisiologia , Técnicas de Cultura de Órgãos , Órgão Espiral/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Estria Vascular/metabolismo
18.
Proc Natl Acad Sci U S A ; 107(5): 2001-6, 2010 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-20080694

RESUMO

Protein ubiquitination controls the cellular fate of numerous eukaryotic proteins. Despite its importance, many fundamental questions remain regarding its mechanism. One such question is how ubiquitination alters the biophysical properties of the modified protein and whether these alterations are significant in the cellular context. In this study, we investigate the effects of ubiquitination on the folding thermodynamics and mechanism of various substrates using computational tools and find that ubiquitination changes the thermal stability of modified proteins in a manner relevant to cellular processes. These changes depend on the substrate modification site and on the type of ubiquitination. Ubiquitination of the substrate Ubc7 at the residues that are modified in vivo prior to proteasomal degradation uniquely results in significant thermal destabilization and a local unwinding near the modification site, which indicates that ubiquitination possibly facilitates the unfolding process and improves substrate degradation efficiency. With respect to the substrate p19(4inkd), our results support a synergetic effect of ubiquitination and phosphorylation on the degradation process via enhanced thermal destabilization. Our study implies that, in addition to its known role as a recognition signal, the ubiquitin attachment may be directly involved in the cellular process it regulates by changing the biophysical properties of the substrate.


Assuntos
Proteínas/química , Proteínas/metabolismo , Ubiquitina/química , Ubiquitina/metabolismo , Fenômenos Biofísicos , Inibidor de Quinase Dependente de Ciclina p19/química , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Modelos Moleculares , Fosforilação , Dobramento de Proteína , Estabilidade Proteica , Termodinâmica , Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação
19.
Proc Natl Acad Sci U S A ; 107(1): 187-92, 2010 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-19966290

RESUMO

Chromosome translocations between Ig (Ig) and non-Ig genes are frequently associated with B-cell lymphomas in humans and mice. The best characterized of these is c-myc/IgH translocation, which is associated with Burkitt's lymphoma. These translocations are caused by activation-induced cytidine deaminase (AID), which produces double-strand DNA breaks in both genes. c-myc/IgH translocations are rare events, in part because ATM, p53, and p19 actively suppress them. To further define the mechanism of protection against the accumulation of cells that bear c-myc/IgH translocation, we assayed B cells from mice that carry mutations in cell-cycle and apoptosis regulator proteins that act downstream of p53. We find that PUMA, Bim, and PKCdelta are required for protection against c-myc/IgH translocation, whereas Bcl-XL and BAFF enhance c-myc/IgH translocation. Whether these effects are general or specific to c-myc/IgH translocation and whether AID produces dsDNA breaks in genes other than c-myc and Ig is not known. To examine these questions, we developed an assay for translocation between IgH and Igbeta, both of which are somatically mutated by AID. Igbeta/IgH, like c-myc/IgH translocations, are AID-dependent, and AID is responsible for lesions on IgH and the non-IgH translocation partners. However, ATM, p53, and p19 do not protect against Igbeta/IgH translocations. Instead, B cells are protected against Igbeta/IgH translocations by a BAFF- and PKCdelta-dependent pathway. We conclude that AID-induced double-strand breaks in non-Ig genes other than c-myc lead to their translocation, and that at least two nonoverlapping pathways protect against translocations in primary B cells.


Assuntos
Citidina Desaminase/metabolismo , Translocação Genética , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Linfócitos B/imunologia , Proteína 11 Semelhante a Bcl-2 , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Transformação Celular Neoplásica , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p19/genética , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Citidina Desaminase/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Genes de Imunoglobulinas , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Hipermutação Somática de Imunoglobulina , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
20.
J Biol Chem ; 286(27): 24079-88, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21566120

RESUMO

We have analyzed the role of the thyroid hormone receptors (TRs) in epidermal homeostasis. Reduced keratinocyte proliferation is found in interfollicular epidermis of mice lacking the thyroid hormone binding isoforms TRα1 and TRß (KO mice). Similar results were obtained in hypothyroid animals, showing the important role of the liganded TRs in epidermal proliferation. In addition, KO and hypothyroid animals display decreased hyperplasia in response to 12-O-tetradecanolyphorbol-13-acetate. Both receptor isoforms play overlapping functional roles in the skin because mice lacking individually TRα1 or TRß also present a proliferative defect but not as marked as that found in double KO mice. Defective proliferation in KO mice is associated with reduction of cyclin D1 expression and up-regulation of the cyclin-dependent kinase inhibitors p19 and p27. Paradoxically, ERK and AKT activity and expression of downstream targets, such as AP-1 components, are increased in KO mice. Increased p65/NF-κB and STAT3 phosphorylation and, as a consequence, augmented expression of chemokines and proinflammatory cytokines is also found in these animals. These results show that thyroid hormones and their receptors are important mediators of skin proliferation and demonstrate that TRs act as endogenous inhibitors of skin inflammation, most likely due to interference with AP-1, NF-κB, and STAT3 activation.


Assuntos
Proliferação de Células , Dermatite/metabolismo , Epiderme/metabolismo , Receptores alfa dos Hormônios Tireóideos/metabolismo , Receptores beta dos Hormônios Tireóideos/metabolismo , Animais , Carcinógenos/farmacologia , Ciclina D1/genética , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p19/genética , Inibidor de Quinase Dependente de Ciclina p19/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Citocinas/genética , Citocinas/metabolismo , Dermatite/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Receptores alfa dos Hormônios Tireóideos/genética , Receptores beta dos Hormônios Tireóideos/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa