Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 74(4): 651-663.e8, 2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-30954402

RESUMO

Accumulating evidence supports the role of the DNA damage response (DDR) in the negative regulation of tumorigenesis. Here, we found that DDR signaling poises a series of epigenetic events, resulting in activation of pro-tumorigenic genes but can go as far as reactivation of the pluripotency gene OCT4. Loss of DNA methylation appears to be a key initiating event in DDR-dependent OCT4 locus reactivation although full reactivation required the presence of a driving oncogene, such as Myc and macroH2A downregulation. Using genetic-lineage-tracing experiments and an in situ labeling approach, we show that DDR-induced epigenetic reactivation of OCT4 regulates the resistance to chemotherapy and contributes to tumor relapse both in mouse and primary human cancers. In turn, deletion of OCT4 reverses chemoresistance and delays the relapse. Here, we uncovered an unexpected tumor-promoting role of DDR in cancer cell reprogramming, providing novel therapeutic entry points for cancer intervention strategies.


Assuntos
Carcinogênese/genética , Metilação de DNA/genética , Neoplasias/genética , Fator 3 de Transcrição de Octâmero/genética , Animais , Reprogramação Celular/genética , Dano ao DNA/genética , Epigênese Genética/genética , Regulação Neoplásica da Expressão Gênica , Histonas/genética , Humanos , Camundongos , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Recidiva , Transdução de Sinais/genética
2.
Circulation ; 147(8): 650-666, 2023 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-36515093

RESUMO

BACKGROUND: Senescent cells (SCs) are involved in proliferative disorders, but their role in pulmonary hypertension remains undefined. We investigated SCs in patients with pulmonary arterial hypertension and the role of SCs in animal pulmonary hypertension models. METHODS: We investigated senescence (p16, p21) and DNA damage (γ-H2AX, 53BP1) markers in patients with pulmonary arterial hypertension and murine models. We monitored p16 activation by luminescence imaging in p16-luciferase (p16LUC/+) knock-in mice. SC clearance was obtained by a suicide gene (p16 promoter-driven killer gene construct in p16-ATTAC mice), senolytic drugs (ABT263 and cell-permeable FOXO4-p53 interfering peptide [FOXO4-DRI]), and p16 inactivation in p16LUC/LUC mice. We investigated pulmonary hypertension in mice exposed to normoxia, chronic hypoxia, or hypoxia+Sugen, mice overexpressing the serotonin transporter (SM22-5-HTT+), and rats given monocrotaline. RESULTS: Patients with pulmonary arterial hypertension compared with controls exhibited high lung p16, p21, and γ-H2AX protein levels, with abundant vascular cells costained for p16, γ-H2AX, and 53BP1. Hypoxia increased thoracic bioluminescence in p16LUC/+ mice. In wild-type mice, hypoxia increased lung levels of senescence and DNA-damage markers, senescence-associated secretory phenotype components, and p16 staining of pulmonary endothelial cells (P-ECs, 30% of lung SCs in normoxia), and pulmonary artery smooth muscle cells. SC elimination by suicide gene or ABT263 increased the right ventricular systolic pressure and hypertrophy index, increased vessel remodeling (higher dividing proliferating cell nuclear antigen-stained vascular cell counts during both normoxia and hypoxia), and markedly decreased lung P-ECs. Pulmonary hemodynamic alterations and lung P-EC loss occurred in older p16LUC/LUC mice, wild-type mice exposed to Sugen or hypoxia+Sugen, and SM22-5-HTT+ mice given either ABT263 or FOXO4-DRI, compared with relevant controls. The severity of monocrotaline-induced pulmonary hypertension in rats was decreased slightly by ABT263 for 1 week but was aggravated at 3 weeks, with loss of P-ECs. CONCLUSIONS: Elimination of senescent P-ECs by senolytic interventions may worsen pulmonary hemodynamics. These results invite consideration of the potential impact on pulmonary vessels of strategies aimed at controlling cell senescence in various contexts.


Assuntos
Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Camundongos , Ratos , Animais , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/metabolismo , Células Endoteliais/metabolismo , Monocrotalina/metabolismo , Senoterapia , Artéria Pulmonar , Hipertensão Pulmonar Primária Familiar/metabolismo , Hipóxia/metabolismo , Senescência Celular , Fatores de Transcrição Forkhead/metabolismo
3.
Genes Dev ; 30(23): 2623-2636, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28007785

RESUMO

Expansion of neoplastic lesions generates the initial signal that instigates the creation of a tumor niche. Nontransformed cell types within the microenvironment continuously coevolve with tumor cells to promote tumorigenesis. Here, we identify p38MAPK as a key component of human lung cancer, and specifically stromal interactomes, which provides an early, protumorigenic signal in the tissue microenvironment. We found that lung cancer growth depends on short-distance cues produced by the cancer niche in a p38-dependent manner. We identified fibroblast-specific hyaluronan synthesis at the center of p38-driven tumorigenesis, which regulates early stromal fibroblast activation, the conversion to carcinoma-associated fibroblasts (CAFs), and cancer cell proliferation. Systemic down-regulation of p38MAPK signaling in a knock-in model with substitution of activating Tyr182 to phenylalanine or conditional ablation of p38 in fibroblasts has a significant tumor-suppressive effect on K-ras lung tumorigenesis. Furthermore, both Kras-driven mouse lung tumors and orthotopically grown primary human lung cancers show a significant sensitivity to both a chemical p38 inhibitor and an over-the-counter inhibitor of hyaluronan synthesis. We propose that p38MAPK-hyaluronan-dependent reprogramming of the tumor microenvironment plays a critical role in driving lung tumorigenesis, while blocking this process could have far-reaching therapeutic implications.


Assuntos
Carcinogênese/genética , Carcinogênese/patologia , Ácido Hialurônico/metabolismo , Neoplasias Pulmonares/fisiopatologia , Microambiente Tumoral/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Antineoplásicos/farmacologia , Proliferação de Células , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Reprogramação Celular/genética , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Fibroblastos , Regulação Neoplásica da Expressão Gênica , Técnicas de Introdução de Genes , Humanos , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
4.
Proc Natl Acad Sci U S A ; 109(2): E68-75, 2012 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-22065775

RESUMO

The inactivation of the p53 tumor suppressor pathway in many cancers often increases their resistance to anticancer therapy. Here we show that a previously proposed strategy directed to Wip1 inhibition could be ineffective in tumors lacking p53. On the contrary, Wip1 overexpression sensitized these tumors to chemotherapeutic agents. This effect was mediated through interaction between Wip1 and RUNX2 that resulted, in response to anticancer treatment, in RUNX2-dependent transcriptional induction of the proapoptotic Bax protein. The potentiating effects of Wip1 overexpression on chemotherapeutic agents were directed only to tumor cells lacking p53. The overexpression of Wip1 in normal tissues provided protection from cisplatin-induced apoptosis through decreased strength of upstream signaling to p53. Thus, Wip1 phosphatase promotes apoptosis in p53-negative tumors and protects normal tissues during treatment with anticancer agents.


Assuntos
Antineoplásicos/farmacologia , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias/tratamento farmacológico , Fosfoproteínas Fosfatases/metabolismo , Proteína Supressora de Tumor p53/deficiência , Animais , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Western Blotting , Linhagem Celular , Primers do DNA/genética , Sinergismo Farmacológico , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Neoplasias/metabolismo , Plasmídeos/genética , Proteína Fosfatase 2C , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/metabolismo
5.
Trends Biochem Sci ; 35(2): 109-14, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19879149

RESUMO

The PP2C family serine/threonine phosphatase WIP1 is characterized by distinctive oncogenic properties mediated by inhibitory functions on several tumor suppressor pathways, including ATM, CHK2, p38MAPK and p53. PPM1D, the gene encoding WIP1, is aberrantly amplified in different types of human primary cancers, and its deletion in mice results in a profound tumor-resistant phenotype. Numerous downstream targets of WIP1 have been identified, and genetic studies confirm that some play a part in tumorigenesis. Recent evidence highlights a new role for WIP1 in the regulation of a cell-autonomous decline in proliferation of certain self-renewing cell types, including pancreatic beta-cells, with advancing age. These emerging functions of WIP1 make it a potent therapeutic target against cancer and aging.


Assuntos
Envelhecimento/fisiologia , Neoplasias/enzimologia , Fosfoproteínas Fosfatases/metabolismo , Animais , Humanos , Neoplasias/metabolismo , Fosfoproteínas Fosfatases/genética , Proteína Fosfatase 2C
6.
Breast Cancer Res ; 15(1): R10, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23369183

RESUMO

INTRODUCTION: The molecular circuitry of different cell types dictates their normal function as well as their response to oncogene activation. For instance, mice lacking the Wip1 phosphatase (also known as PPM1D; protein phosphatase magnesium-dependent 1D) have a delay in HER2/neu (human epidermal growth factor 2), but not Wnt1-induced mammary tumor formation. This suggests a cell type-specific reliance on Wip1 for tumorigenesis, because alveolar progenitor cells are the likely target for transformation in the MMTV(mouse mammary tumor virus)-neu but not MMTV-wnt1 breast cancer model. METHODS: In this study, we used the Wip1-knockout mouse to identify the cell types that are dependent on Wip1 expression and therefore may be involved in the early stages of HER2/neu-induced tumorigenesis. RESULTS: We found that alveolar development during pregnancy was reduced in Wip1-knockout mice; however, this was not attributable to changes in alveolar cells themselves. Unexpectedly, Wip1 allows steroid hormone-receptor-positive cells but not alveolar progenitors to activate STAT5 (signal transducer and activator of transcription 5) in the virgin state. In the absence of Wip1, hormone-receptor-positive cells have significantly reduced transcription of RANKL (receptor activator of nuclear factor kappa-B ligand) and IGF2 (insulin-like growth factor 2), paracrine stimulators of alveolar development. In the MMTV-neu model, HER2/neu activates STAT5 in alveolar progenitor cells independent of Wip1, but HER2/neu does not override the defect in STAT5 activation in Wip1-deficient hormone-sensing cells, and paracrine stimulation remains attenuated. Moreover, ERK (extracellular signal-regulated kinase) activation by HER2/neu in hormone-sensing cells is also Wip1 dependent. CONCLUSIONS: We identified Wip1 as a potentiator of prolactin and HER2/neu signaling strictly in the molecular context of hormone-sensing cells. Furthermore, our findings highlight that hormone-sensing cells convert not only estrogen and progesterone but also prolactin signals into paracrine instructions for mammary gland development. The instructive role of hormone-sensing cells in premalignant development suggests targeting Wip1 or prolactin signaling as an orthogonal strategy for inhibiting breast cancer development or relapse.


Assuntos
Neoplasias da Mama/genética , Transformação Celular Neoplásica , Neoplasias Mamárias Animais/genética , Fosfoproteínas Fosfatases/genética , Animais , Neoplasias da Mama/patologia , Estrogênios/metabolismo , Feminino , Humanos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Knockout , Fosfoproteínas Fosfatases/metabolismo , Gravidez , Prolactina/metabolismo , Proteína Fosfatase 2C , Receptor ErbB-2/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais
7.
Nat Genet ; 36(4): 343-50, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14991053

RESUMO

Modulation of tumor suppressor activities may provide new opportunities for cancer therapy. Here we show that disruption of the gene Ppm1d encoding Wip1 phosphatase activated the p53 and p16 (also called Ink4a)-p19 (also called ARF) pathways through p38 MAPK signaling and suppressed in vitro transformation of mouse embryo fibroblasts (MEFs) by oncogenes. Disruption of the gene Cdkn2a (encoding p16 and p19), but not of Trp53 (encoding p53), reconstituted cell transformation in Ppm1d-null MEFs. In vivo, deletion of Ppm1d in mice bearing mouse mammary tumor virus (MMTV) promoter-driven oncogenes Erbb2 (also called c-neu) or Hras1 impaired mammary carcinogenesis, whereas reduced expression of p16 and p19 by methylation-induced silencing or inactivation of p38 MAPK correlated with tumor appearance. We conclude that inactivation or depletion of the Wip1 phosphatase with resultant p38 MAPK activation suppresses tumor appearance by modulating the Cdkn2a tumor-suppressor locus.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Neoplasias Mamárias Experimentais/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Fosfoproteínas Fosfatases/antagonistas & inibidores , Proteína Supressora de Tumor p14ARF/metabolismo , Animais , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteína Fosfatase 2C , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
8.
Nat Genet ; 31(2): 210-5, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12021785

RESUMO

Expression of oncogenic Ras in primary human cells activates p53, thereby protecting cells from transformation. We show that in Ras-expressing IMR-90 cells, p53 is phosphorylated at Ser33 and Ser46 by the p38 mitogen-activated protein kinase (MAPK). Activity of p38 MAPK is regulated by the p53-inducible phosphatase PPM1D, creating a potential feedback loop. Expression of oncogenic Ras suppresses PPM1D mRNA induction, leaving p53 phosphorylated at Ser33 and Ser46 and in an active state. Retrovirus-mediated overexpression of PPM1D reduced p53 phosphorylation at these sites, abrogated Ras-induced apoptosis and partially rescued cells from cell-cycle arrest. Inactivation of p38 MAPK (the product of Mapk14) in vivo by gene targeting or by PPM1D overexpression expedited tumor formation after injection of mouse embryo fibroblasts (MEFs) expressing E1A+Ras into nude mice. The gene encoding PPM1D (PPM1D, at 17q22/q23) is amplified in human breast-tumor cell lines and in approximately 11% of primary breast tumors, most of which harbor wildtype p53. These findings suggest that inactivation of the p38 MAPK through PPM1D overexpression resulting from PPM1D amplification contributes to the development of human cancers by suppressing p53 activation.


Assuntos
Neoplasias da Mama/genética , Cromossomos Humanos Par 17 , Amplificação de Genes , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas de Neoplasias , Fosfoproteínas Fosfatases/genética , Proteína Supressora de Tumor p53/genética , Animais , Neoplasias da Mama/etiologia , Feminino , Fibroblastos/fisiologia , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Fosforilação , Proteína Fosfatase 2C , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno
9.
Nat Cell Biol ; 25(9): 1265-1278, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37652981

RESUMO

Despite advances in four-factor (4F)-induced reprogramming (4FR) in vitro and in vivo, how 4FR interconnects with senescence remains largely under investigated. Here, using genetic and chemical approaches to manipulate senescent cells, we show that removal of p16High cells resulted in the 4FR of somatic cells into totipotent-like stem cells. These cells expressed markers of both pluripotency and the two-cell embryonic state, readily formed implantation-competent blastoids and, following morula aggregation, contributed to embryonic and extraembryonic lineages. We identified senescence-dependent regulation of nicotinamide N-methyltransferase as a key mechanism controlling the S-adenosyl-L-methionine levels during 4FR that was required for expression of the two-cell genes and acquisition of an extraembryonic potential. Importantly, a partial 4F epigenetic reprogramming in old mice was able to reverse several markers of liver aging only in conjunction with the depletion of p16High cells. Our results show that the presence of p16High senescent cells limits cell plasticity, whereas their depletion can promote a totipotent-like state and histopathological tissue rejuvenation during 4F reprogramming.


Assuntos
Plasticidade Celular , Reprogramação Celular , Animais , Camundongos , Reprogramação Celular/genética , Envelhecimento/genética , Implantação do Embrião , Epigenômica
10.
J Exp Med ; 203(13): 2793-9, 2006 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-17158963

RESUMO

The ataxia telangiectasia mutated (ATM) kinase is a key tumor suppressor that regulates numerous cell cycle checkpoints as well as apoptosis. Here, we report that ATM is a critical player in the regulation of apoptosis and lymphomagenesis in the presence of c-myc. In turn, deletion of the inhibitory ATM phosphatase, Wip1, results in ATM up-regulation and suppression of Emicro-myc-induced B cell lymphomas. Using mouse genetic crosses, we show that the onset of myc-induced lymphomas is dramatically delayed in Wip1-null mice in an ATM- and p53-, but not p38 MAPK- or Arf-, dependent manner. We propose that Wip1 phosphatase is critical for regulating the ATM-mediated tumor surveillance network.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Linfoma/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose/genética , Proteínas Mutadas de Ataxia Telangiectasia , Linfócitos B/metabolismo , Western Blotting , Inibidores de Caspase , Caspases/metabolismo , Proteínas de Ciclo Celular/genética , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/genética , Expressão Gênica , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/metabolismo , Linfoma/genética , Linfoma/patologia , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Proteínas de Neoplasias/genética , Fosfoproteínas Fosfatases/genética , Fosforilação , Ligação Proteica , Proteína Fosfatase 2C , Proteínas Serina-Treonina Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Nat Cell Biol ; 5(6): 545-51, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12766774

RESUMO

Negative regulation of the Cdc25C protein phosphatase by phosphorylation on Ser 216, the 14-3-3-binding site, is an important regulatory mechanism used by cells to block mitotic entry under normal conditions and after DNA damage. During mitosis, Cdc25C is not phosphorylated on Ser 216 and ionizing radiation (IR) does not induce either phosphorylation of Ser 216, or binding to 14-3-3. Here, we show that Cdc25C is phosphorylated on Ser 214 during mitosis, which in turn prevents phosphorylation of Ser 216. Mutation of Ser 214 to Ala reconstitutes Ser 216 phosphorylation and 14-3-3 binding during mitosis. Introduction of exogenous Cdc25C(S214A) into HeLa cells depleted of endogenous Cdc25C results in a substantial delay to mitotic entry. This effect was fully reversed in a S214A/S216A double-mutant, implying that the inhibitory effect of S214A mutant was entirely dependent on Ser 216 phosphorylation. A similar regulatory mechanism may also apply to another mitotic phosphatase, Cdc25B, as well as mitotic phosphatases of other species, including Xenopus laevis. We propose that this pathway ensures that Cdc2 remains active once mitosis is initiated and is a key control mechanism for maintaining the proper order of cell-cycle transitions.


Assuntos
Mitose , Fosfatases cdc25/metabolismo , Proteínas 14-3-3 , Alanina/metabolismo , Substituição de Aminoácidos , Antineoplásicos/farmacologia , Dano ao DNA , Fase G2/fisiologia , Células HeLa , Humanos , Mutagênese Sítio-Dirigida , Nocodazol/farmacologia , Fosforilação , Mutação Puntual , Ligação Proteica , Radiação Ionizante , Serina/metabolismo , Tirosina 3-Mono-Oxigenase/química , Fosfatases cdc25/genética
12.
Stem Cells ; 27(6): 1433-42, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19489034

RESUMO

Continual generation of new neural cells from adult neural stem/progenitor cells (NPCs) is an important component of life-long brain plasticity. However, the intrinsic regulation of this process remains poorly defined. Here we report that Wip1 phosphatase, previously studied in oncogenesis, functions as a crucial physiological regulator in adult neural cell generation. Wip1 deficiency resulted in a 90% decrease in new cell formation in adult olfactory bulb, accompanied by aberrantly decreased NPC amplification, stem cell frequency, and self-renewal. At a cellular level, Wip1 knockout NPCs exhibit a prolonged cell cycle, an accumulation at G(2) to M phase transition, and enhanced p53 activity. Interestingly, the impaired M-phase entry and NPC amplification of Wip1-null mice can be reversed in Wip1/p53 double-null mice. Importantly, there is no difference in NPC amplification between p53-null and Wip1/p53 double-null mice. Our data demonstrate that Wip1 regulates the generation of new neural cells in adult olfactory bulb specifically through p53-dependent M-phase entry of the NPC cell cycle.


Assuntos
Células-Tronco Adultas/citologia , Neurogênese/fisiologia , Neurônios/citologia , Bulbo Olfatório/citologia , Fosfoproteínas Fosfatases/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Células-Tronco Adultas/metabolismo , Animais , Apoptose/fisiologia , Western Blotting , Ciclo Celular , Diferenciação Celular/genética , Citometria de Fluxo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Bulbo Olfatório/metabolismo , Fosfoproteínas Fosfatases/genética , Proteína Fosfatase 2C , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Cell Biol ; 169(5): 755-63, 2005 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-15928203

RESUMO

Lymphocytes are the central mediators of the immune response, requiring cytokines for survival and proliferation. Survival signaling targets the Bcl-2 family of apoptotic mediators, however, the pathway for the cytokine-driven proliferation of lymphocytes is poorly understood. Here we show that cytokine-induced cell cycle progression is not solely dependent on the synthesis of cyclin-dependent kinases (Cdks) or cyclins. Rather, we observe that in lymphocyte cell lines dependent on interleukin-3 or interleukin-7, or primary lymphocytes dependent on interleukin 7, the phosphatase Cdc25A is the critical mediator of proliferation. Withdrawal of IL-7 or IL-3 from dependent lymphocytes activates the stress kinase, p38 MAPK, which phosphorylates Cdc25A, inducing its degradation. As a result, Cdk/cyclin complexes remain phosphorylated and inactive and cells arrest before the induction of apoptosis. Inhibiting p38 MAPK or expressing a mutant Cdc25A, in which the two p38 MAPK target sites, S75 and S123, are altered, renders cells resistant to cytokine withdrawal, restoring the activity of Cdk/cyclin complexes and driving the cell cycle independent of a growth stimulus.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Citocinas/imunologia , Imunidade/imunologia , Linfócitos/metabolismo , Fosfatases cdc25/metabolismo , Animais , Apoptose/fisiologia , Domínio Catalítico/fisiologia , Proteínas de Ciclo Celular/imunologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Citocinas/metabolismo , Interleucina-3/imunologia , Interleucina-3/farmacologia , Interleucina-7/imunologia , Interleucina-7/farmacologia , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Camundongos , Camundongos Knockout , Mutação/fisiologia , Fosforilação , Transdução de Sinais/imunologia , Estresse Fisiológico/imunologia , Estresse Fisiológico/metabolismo , Fosfatases cdc25/genética , Fosfatases cdc25/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Aging (Albany NY) ; 12(11): 11152-11160, 2020 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-32535553

RESUMO

Data obtained from genetically modified mouse models suggest a detrimental role for p16High senescent cells in physiological aging and age-related pathologies. Our recent analysis of aging mice revealed a continuous and noticeable accumulation of liver sinusoid endothelial cells (LSECs) expressing numerous senescence markers, including p16. At early stage, senescent LSECs show an enhanced ability to clear macromolecular waste and toxins including oxidized LDL (oxLDL). Later in life, however, the efficiency of this important detoxifying function rapidly declines potentially due to increased endothelial thickness and senescence-induced silencing of scavenger receptors and endocytosis genes. This inability to detoxify toxins and macromolecular waste, which can be further exacerbated by increased intestinal leakiness with age, might be an important contributing factor to animal death. Here, we propose how LSEC senescence could serve as an endogenous clock that ultimately controls longevity and outline some of the possible approaches to extend the lifespan.


Assuntos
Envelhecimento/fisiologia , Senescência Celular/fisiologia , Células Endoteliais/metabolismo , Fígado/citologia , Animais , Lipoproteínas LDL/metabolismo , Fígado/metabolismo , Longevidade/fisiologia , Camundongos , Modelos Animais
15.
Cell Metab ; 32(1): 87-99.e6, 2020 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-32485135

RESUMO

The accumulation of senescent cells can drive many age-associated phenotypes and pathologies. Consequently, it has been proposed that removing senescent cells might extend lifespan. Here, we generated two knockin mouse models targeting the best-characterized marker of senescence, p16Ink4a. Using a genetic lineage tracing approach, we found that age-induced p16High senescence is a slow process that manifests around 10-12 months of age. The majority of p16High cells were vascular endothelial cells mostly in liver sinusoids (LSECs), and to lesser extent macrophages and adipocytes. In turn, continuous or acute elimination of p16High senescent cells disrupted blood-tissue barriers with subsequent liver and perivascular tissue fibrosis and health deterioration. Our data show that senescent LSECs are not replaced after removal and have important structural and functional roles in the aging organism. In turn, delaying senescence or replacement of senescent LSECs could represent a powerful tool in slowing down aging.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Envelhecimento/metabolismo , Animais , Células Cultivadas , Senescência Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Células Endoteliais/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
16.
Cell Metab ; 29(1): 124-140.e10, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30293773

RESUMO

Dysregulation of extracellular matrix (ECM) deposition and cellular metabolism promotes tumor aggressiveness by sustaining the activity of key growth, invasion, and survival pathways. Yet mechanisms by which biophysical properties of ECM relate to metabolic processes and tumor progression remain undefined. In both cancer cells and carcinoma-associated fibroblasts (CAFs), we found that ECM stiffening mechanoactivates glycolysis and glutamine metabolism and thus coordinates non-essential amino acid flux within the tumor niche. Specifically, we demonstrate a metabolic crosstalk between CAF and cancer cells in which CAF-derived aspartate sustains cancer cell proliferation, while cancer cell-derived glutamate balances the redox state of CAFs to promote ECM remodeling. Collectively, our findings link mechanical stimuli to dysregulated tumor metabolism and thereby highlight a new metabolic network within tumors in which diverse fuel sources are used to promote growth and aggressiveness. Furthermore, this study identifies potential metabolic drug targets for therapeutic development in cancer.


Assuntos
Ácido Aspártico/metabolismo , Neoplasias da Mama/metabolismo , Fibroblastos Associados a Câncer/metabolismo , Carcinoma/metabolismo , Ácido Glutâmico/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Fibroblastos Associados a Câncer/patologia , Linhagem Celular , Matriz Extracelular , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP
17.
Curr Opin Genet Dev ; 12(1): 92-7, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11790561

RESUMO

The mechanism controlling G(2)/M checkpoint activation after DNA damage was thought to be mediated primarily by nuclear Chk1/Chk2 kinases. Recent evidence indicates that this checkpoint is more complex, involving at least two different biochemical systems that target the Cdc25B and Cdc25C phosphatases. Following genotoxic stress, different kinases integrate signaling from the damaged DNA and other damaged cellular components to regulate Cdc25 inactivation. Our current model for G(2)/M checkpoint activation after genotoxic stress is discussed emphasizing the roles for Chk1 and p38 kinases in checkpoint regulation.


Assuntos
Fase G2 , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Mitose , Proteínas Quinases/fisiologia , Animais , Proteínas de Ciclo Celular/metabolismo , Quinase 1 do Ponto de Checagem , Dano ao DNA , Humanos , Proteínas Quinases Ativadas por Mitógeno/química , Fosforilação , Proteínas Quinases/química , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno
18.
Cancer Res ; 66(6): 2928-36, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16540640

RESUMO

Activation of apoptosis is believed to be critical for the role of p53 as a tumor suppressor. Here, we report a new mouse strain carrying a human p53 transgene in the mouse p53-null background. Expression of human p53 in these mice was comparable with wild-type murine p53; however, transactivation, induction of apoptosis, and G(1)-S checkpoint, but not transrepression or regulation of a centrosomal checkpoint, were deregulated. Although multiple functions of p53 were abrogated, mice carrying the human p53 transgene did not show early onset of tumors as typically seen for p53-null mice. In contrast, human p53 in the p53-null background did not prevent accelerated tumor development after genotoxic or oncogenic stress. Such behavior of human p53 expressed at physiologic levels in transgenic cells could be explained by unexpectedly high binding with Mdm2. By using Nutlin-3a, an inhibitor of the interaction between Mdm2 and p53, we were able to partially reconstitute p53 transactivation and apoptosis in transgenic cells. Our findings indicate that the interaction between p53 and Mdm2 controls p53 transcriptional activity in homeostatic tissues and regulates DNA damage- and oncogene-induced, but not spontaneous, tumorigenesis.


Assuntos
Apoptose/genética , Genes p53 , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Transgenes , Proteína Supressora de Tumor p53/biossíntese , Animais , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteína Supressora de Tumor p53/genética
19.
Cancer Res ; 78(18): 5229-5242, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30026329

RESUMO

In squamous cell carcinoma (SCC), tissue invasion by collectively invading cells requires physical forces applied by tumor cells on their surrounding extracellular matrix (ECM). Cancer-related ECM is composed of thick collagen bundles organized by carcinoma-associated fibroblasts (CAF) within the tumor stroma. Here, we show that SCC cell collective invasion is driven by the matrix-dependent mechano-sensitization of EGF signaling in cancer cells. Calcium (Ca2+) was a potent intracellular second messenger that drove actomyosin contractility. Tumor-derived matrix stiffness and EGFR signaling triggered increased intracellular Ca2+ through CaV1.1 expression in SCC cells. Blocking L-type calcium channel expression or activity using Ca2+ channel blockers verapamil and diltiazem reduced SCC cell collective invasion both in vitro and in vivo These results identify verapamil and diltiazem, two drugs long used in medical care, as novel therapeutic strategies to block the tumor-promoting activity of the tumor niche.Significance: This work demonstrates that calcium channels blockers verapamil and diltiazem inhibit mechano-sensitization of EGF-dependent cancer cell collective invasion, introducing potential clinical strategies against stromal-dependent collective invasion.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/18/5229/F1.large.jpg Cancer Res; 78(18); 5229-42. ©2018 AACR.


Assuntos
Sinalização do Cálcio , Carcinoma de Células Escamosas/patologia , Matriz Extracelular/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Actomiosina/metabolismo , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Canais de Cálcio Tipo L , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Colágeno/metabolismo , Diltiazem/farmacologia , Receptores ErbB/metabolismo , Fibroblastos/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Invasividade Neoplásica , Esferoides Celulares , Verapamil/farmacologia
20.
Mol Cell Biol ; 23(11): 3859-71, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12748288

RESUMO

The activation of p53 is a guardian mechanism to protect primary cells from malignant transformation; however, the details of the activation of p53 by oncogenic stress are still incomplete. In this report we show that in Gadd45a(-/-) mouse embryo fibroblasts (MEF), overexpression of H-ras activates extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) but not p38 kinase, and this correlates with the loss of H-ras-induced cell cycle arrest (premature senescence). Inhibition of p38 mitogen-activated protein kinase (MAPK) activation correlated with the deregulation of p53 activation, and both a p38 MAPK chemical inhibitor and the expression of a dominant-negative p38alpha inhibited p53 activation in the presence of H-ras in wild-type MEF. p38, but not ERK or JNK, was found in a complex with Gadd45 proteins. The region of interaction was mapped to amino acids 71 to 96, and the central portion (amino acids 71 to 124) of Gadd45a was required for p38 MAPK activation in the presence of H-ras. Our results indicate that this Gadd45/p38 pathway plays an important role in preventing oncogene-induced growth at least in part by regulating the p53 tumor suppressor.


Assuntos
Proteínas de Ciclo Celular , Ciclo Celular/fisiologia , Genes ras , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Nucleares/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Senescência Celular/fisiologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , Fibroblastos/citologia , Fibroblastos/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Ligação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA