Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO Rep ; 19(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29420235

RESUMEN

Caseinolytic peptidase P (ClpP) is a mammalian quality control protease that is proposed to play an important role in the initiation of the mitochondrial unfolded protein response (UPRmt), a retrograde signaling response that helps to maintain mitochondrial protein homeostasis. Mitochondrial dysfunction is associated with the development of metabolic disorders, and to understand the effect of a defective UPRmt on metabolism, ClpP knockout (ClpP-/-) mice were analyzed. ClpP-/- mice fed ad libitum have reduced adiposity and paradoxically improved insulin sensitivity. Absence of ClpP increased whole-body energy expenditure and markers of mitochondrial biogenesis are selectively up-regulated in the white adipose tissue (WAT) of ClpP-/- mice. When challenged with a metabolic stress such as high-fat diet, despite similar caloric intake, ClpP-/- mice are protected from diet-induced obesity, glucose intolerance, insulin resistance, and hepatic steatosis. Our results show that absence of ClpP triggers compensatory responses in mice and suggest that ClpP might be dispensable for mammalian UPRmt initiation. Thus, we made an unexpected finding that deficiency of ClpP in mice is metabolically beneficial.


Asunto(s)
Endopeptidasa Clp/genética , Resistencia a la Insulina/genética , Mitocondrias/genética , Obesidad/genética , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Obesidad/metabolismo , Obesidad/patología , Respuesta de Proteína Desplegada/genética
2.
Breast Cancer Res Treat ; 150(3): 487-99, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25788226

RESUMEN

Triple-negative breast cancer (TNBC), the most aggressive breast cancer subtype, occurs in younger women and is associated with poor prognosis. Gain-of-function mutations in TP53 are a frequent occurrence in TNBC and have been demonstrated to repress apoptosis and up-regulate cell cycle progression. Even though TNBC responds to initial chemotherapy, resistance to chemotherapy develops and is a major clinical problem. Tumor recurrence eventually occurs and most patients die from their disease. An urgent need exists to identify molecular-targeted therapies that can enhance chemotherapy response. In the present study, we report that targeting PELP1, an oncogenic co-regulator molecule, could enhance the chemotherapeutic response of TNBC through the inhibition of cell cycle progression and activation of apoptosis. We demonstrate that PELP1 interacts with MTp53, regulates its recruitment, and alters epigenetic marks at the target gene promoters. PELP1 knockdown reduced MTp53 target gene expression, resulting in decreased cell survival and increased apoptosis upon genotoxic stress. Mechanistic studies revealed that PELP1 depletion contributes to increased stability of E2F1, a transcription factor that regulates both cell cycle and apoptosis in a context-dependent manner. Further, PELP1 regulates E2F1 stability in a KDM1A-dependent manner, and PELP1 phosphorylation at the S1033 residue plays an important role in mediating its oncogenic functions in TNBC cells. Accordingly, depletion of PELP1 increased the expression of E2F1 target genes and reduced TNBC cell survival in response to genotoxic agents. PELP1 phosphorylation was significantly greater in the TNBC tumors than in the other subtypes of breast cancer and in the normal tissues. These findings suggest that PELP1 is an important molecular target in TNBC, and that PELP1-targeted therapies may enhance response to chemotherapies.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Factor de Transcripción E2F1/metabolismo , Mutación , Factores de Transcripción/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Proteína p53 Supresora de Tumor/genética , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Proteínas Co-Represoras/antagonistas & inhibidores , Proteínas Co-Represoras/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Fosforilación , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Proteína p53 Supresora de Tumor/metabolismo
3.
J Biol Chem ; 287(41): 34225-33, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-22902628

RESUMEN

DNA damage-induced proliferating cell nuclear antigen (PCNA) ubiquitination serves as the key event mediating post-replication repair. Post-replication repair involves either translesion synthesis (TLS) or damage avoidance via template switching. In this study, we have identified and characterized C1orf124 as a regulator of TLS. C1orf124 co-localizes and interacts with unmodified and mono-ubiquitinated PCNA at UV light-induced damage sites, which require the PIP box and UBZ domain of C1orf124. C1orf124 also binds to the AAA-ATPase valosin-containing protein via its SHP domain, and cellular resistance to UV radiation mediated by C1orf124 requires its interactions with valosin-containing protein and PCNA. Interestingly, C1orf124 binds to replicative DNA polymerase POLD3 and PDIP1 under normal conditions but preferentially associates with TLS polymerase η (POLH) upon UV damage. Depletion of C1orf124 compromises PCNA monoubiquitination, RAD18 chromatin association, and RAD18 localization to UV damage sites. Thus, C1orf124 acts at multiple steps in TLS, stabilizes RAD18 and ubiquitinated PCNA at damage sites, and facilitates the switch from replicative to TLS polymerase to bypass DNA lesion.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Ubiquitinación , ADN Polimerasa III/genética , ADN Polimerasa III/metabolismo , Proteínas de Unión al ADN/genética , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Proteína FUS de Unión a ARN/genética , Ubiquitina-Proteína Ligasas , Rayos Ultravioleta/efectos adversos
4.
EMBO Rep ; 11(6): 438-44, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20448663

RESUMEN

Histone methylation has a key role in oestrogen receptor (ERalpha)-mediated transactivation of genes. Proline glutamic acid and leucine-rich protein 1 (PELP1) is a new proto-oncogene that functions as an ERalpha co-regulator. In this study, we identified histone lysine demethylase, KDM1, as a new PELP1-interacting protein. These proteins, PELP1 and KDM1, were both recruited to ERalpha target genes, and PELP1 depletion affected the dimethyl histone modifications at ERalpha target genes. Dimethyl-modified histones H3K4 and H3K9 are recognized by PELP1, and PELP1 alters the substrate specificity of KDM1 from H3K4 to H3K9. Effective demethylation of dimethyl H3K9 by KDM1 requires a KDM1-ERalpha-PELP1 functional complex. These results suggest that PELP1 is a reader of H3 methylation marks and has a crucial role in modulating the histone code at the ERalpha target genes.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Histona Demetilasas/metabolismo , Histonas/metabolismo , Transactivadores/metabolismo , Activación Transcripcional , Línea Celular Tumoral , Proteínas Co-Represoras , Estradiol/farmacología , Humanos , Metilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Proto-Oncogenes Mas , Especificidad por Sustrato/efectos de los fármacos , Factores de Transcripción , Activación Transcripcional/efectos de los fármacos
5.
Breast Cancer Res ; 13(3): R80, 2011 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-21834972

RESUMEN

INTRODUCTION: Current clinical strategies for treating hormonal breast cancer involve the use of anti-estrogens that block estrogen receptor (ER)α functions and aromatase inhibitors that decrease local and systemic estrogen production. Both of these strategies improve outcomes for ERα-positive breast cancer patients, however, development of therapy resistance remains a major clinical problem. Divergent molecular pathways have been described for this resistant phenotype and interestingly, the majority of downstream events in these resistance pathways converge upon the modulation of cell cycle regulatory proteins including aberrant activation of cyclin dependent kinase 2 (CDK2). In this study, we examined whether the CDK inhibitor roscovitine confers a tumor suppressive effect on therapy-resistant breast epithelial cells. METHODS: Using various in vitro and in vivo assays, we tested the effect of roscovitine on three hormonal therapy-resistant model cells: (a) MCF-7-TamR (acquired tamoxifen resistance model); (b) MCF-7-LTLTca (acquired letrozole resistance model); and (c) MCF-7-HER2 that exhibit tamoxifen resistance (ER-growth factor signaling cross talk model). RESULTS: Hormonal therapy-resistant cells exhibited aberrant activation of the CDK2 pathway. Roscovitine at a dose of 20 µM significantly inhibited the cell proliferation rate and foci formation potential of all three therapy-resistant cells. The drug treatment substantially increased the proportion of cells in G2/M cell cycle phase with decreased CDK2 activity and promoted low cyclin D1 levels. Interestingly, roscovitine also preferentially down regulated the ERα isoform and ER-coregulators including AIB1 and PELP1. Results from xenograft studies further showed that roscovitine can attenuate growth of therapy-resistant tumors in vivo. CONCLUSIONS: Roscovitine can reduce cell proliferation and survival of hormone therapy-resistant breast cancer cells. Our results support the emerging concept that inhibition of CDK2 activity has the potential to abrogate growth of hormonal therapy-resistant cells.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Purinas/farmacología , Animales , Antineoplásicos Hormonales/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas Co-Represoras/biosíntesis , Proteínas Co-Represoras/efectos de los fármacos , Ciclina D1/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Coactivador 3 de Receptor Nuclear/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Roscovitina , Factores de Transcripción/biosíntesis , Factores de Transcripción/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Breast Cancer Res Treat ; 130(2): 377-85, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21184269

RESUMEN

The estrogen receptor (ER) is implicated in the progression of breast cancer. Despite positive effects of hormonal therapy, initial or acquired resistance to endocrine therapies frequently occurs. Recent studies suggested ERα-coregulator PELP1 and growth factor receptor ErbB2/HER2 play an essential role in hormonal therapy responsiveness. Src axis couples ERα with HER2 and PELP1, thus representing a new pathway for targeted therapy resistance. To establish the significance of ER-Src axis in PELP1 and HER2 mediated therapy resistance, we have generated model cells that stably express Src-shRNA under conditions of PELP1, HER2 deregulation. Depletion of Src using shRNA substantially reduced E2 mediated activation of Src and MAPK activation in resistant model cells. Pharmacological inhibition of Src using dasatinib, an orally available inhibitor substantially inhibited the growth of therapy resistant MCF7-PELP1, MCF7-HER2, and MCF7-Tam model cells in proliferation assays. In post-menopausal xenograft based studies, treatment with dasatinib significantly inhibited the growth of therapy resistant cells. IHC analysis revealed that the tumors were ERα positive, and dasatinib treated tumors exhibited alterations in Src and MAPK signaling pathways. Combinatorial therapy of tamoxifen with dasatinib showed better therapeutic effect compared to single agent therapy on the growth of therapy resistant PELP1 driven tumors. The results from our study showed that ER-Src axis play an important role in promoting hormonal resistance by proto-oncogenes such as HER2, PELP1, and blocking this axis prevents the development of hormonal independence in vivo. Since PELP1, HER2, and Src kinase are commonly deregulated in breast cancers, combination therapies using both endocrine agents and dasatinib may have better therapeutic effect by delaying the development of hormonal resistance.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Receptor alfa de Estrógeno/metabolismo , Pirimidinas/farmacología , Tiazoles/farmacología , Familia-src Quinasas/metabolismo , Animales , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Dasatinib , Sinergismo Farmacológico , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Letrozol , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Nitrilos/farmacología , Nitrilos/uso terapéutico , Fosforilación , Pirimidinas/uso terapéutico , Interferencia de ARN , Receptor ErbB-2/metabolismo , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Tiazoles/uso terapéutico , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Triazoles/farmacología , Triazoles/uso terapéutico , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/genética
7.
Onco Targets Ther ; 14: 3037-3049, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33994796

RESUMEN

BACKGROUND: Crizotinib inhibits ALK, MET and ROS1 tyrosine kinases but the development of resistance to monotherapy is an issue. The anti-angiogenic properties of pazopanib could overcome crizotinib drug resistance. Additionally, the anti-angiogenic properties of crizotinib could augment the clinical efficacy of pazopanib. METHODS: We evaluated the safety and responses in patients with advanced solid tumors treated with crizotinib and pazopanib. RESULTS: Eighty-two patients (median age 53 years, range 18-78 years) were enrolled. The median number of prior systemic therapies was 3 (range, 0-8). We were able to dose escalate to dose level 8 (crizotinib 250 mg twice daily and pazopanib 800 mg daily) with no MTD identified. Grade 3 or 4 toxicities were seen in 32% of patients with the highest prevalence being fatigue (n=9, 11%), diarrhea (n=6, 7%), vomiting (n=3, 4%), anemia (n=2, 2%) and ALT increased (n=2, 2%). Of the 82 patients, 61 (74%) had measurable disease by RECISTv1.1 and reached first restaging (6 weeks). Partial response (PR) was observed in 6/61 (10%) patients, and stable disease (SD) lasting ≥6 months was observed in 10/61 patients (16%) (total = 16/61 (26%) of patients with SD ≥6 months/PR). CONCLUSION: Dose level 6 (crizotinib 200 mg twice daily and pazopanib 600 mg daily) was the most tolerable dosing of the combination and can be used in future studies. We also observed moderate clinical activity in patients with advanced solid tumors that had received numerous prior therapies.

8.
Clin Cancer Res ; 27(1): 34-42, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33082208

RESUMEN

PURPOSE: The RET proto-oncogene encodes a receptor tyrosine kinase that is activated by gene fusion in 1%-2% of non-small cell lung cancers (NSCLC) and rarely in other cancer types. Selpercatinib is a highly selective RET kinase inhibitor that has recently been approved by the FDA in lung and thyroid cancers with activating RET gene fusions and mutations. Molecular mechanisms of acquired resistance to selpercatinib are poorly understood. PATIENTS AND METHODS: We studied patients treated on the first-in-human clinical trial of selpercatinib (NCT03157129) who were found to have MET amplification associated with resistance to selpercatinib. We validated MET activation as a targetable mediator of resistance to RET-directed therapy, and combined selpercatinib with the MET/ALK/ROS1 inhibitor crizotinib in a series of single patient protocols (SPP). RESULTS: MET amplification was identified in posttreatment biopsies in 4 patients with RET fusion-positive NSCLC treated with selpercatinib. In at least one case, MET amplification was clearly evident prior to therapy with selpercatinib. We demonstrate that increased MET expression in RET fusion-positive tumor cells causes resistance to selpercatinib, and this can be overcome by combining selpercatinib with crizotinib. Using SPPs, selpercatinib with crizotinib were given together generating anecdotal evidence of clinical activity and tolerability, with one response lasting 10 months. CONCLUSIONS: Through the use of SPPs, we were able to offer combination therapy targeting MET-amplified resistance identified on the first-in-human study of selpercatinib. These data suggest that MET dependence is a recurring and potentially targetable mechanism of resistance to selective RET inhibition in advanced NSCLC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-ret/antagonistas & inhibidores , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas , Ensayos Clínicos Fase I como Asunto , Crizotinib/farmacología , Crizotinib/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Amplificación de Genes , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/genética , Proyectos Piloto , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-ret/genética , Pirazoles/farmacología , Pirazoles/uso terapéutico , Piridinas/farmacología , Piridinas/uso terapéutico , Resultado del Tratamiento
9.
Neurooncol Adv ; 1(1): vdz042, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32309805

RESUMEN

BACKGROUND: Glioblastoma (GBM) is a deadly neoplasm of the central nervous system. The molecular mechanisms and players that contribute to GBM development is incompletely understood. METHODS: The expression of PELP1 in different grades of glioma and normal brain tissues was analyzed using immunohistochemistry on a tumor tissue array. PELP1 expression in established and primary GBM cell lines was analyzed by Western blotting. The effect of PELP1 knockdown was studied using cell proliferation, colony formation, migration, and invasion assays. Mechanistic studies were conducted using RNA-seq, RT-qPCR, immunoprecipitation, reporter gene assays, and signaling analysis. Mouse orthotopic models were used for preclinical evaluation of PELP1 knock down. RESULTS: Nuclear receptor coregulator PELP1 is highly expressed in gliomas compared to normal brain tissues, with the highest expression in GBM. PELP1 expression was elevated in established and patient-derived GBM cell lines compared to normal astrocytes. Knockdown of PELP1 resulted in a significant decrease in cell viability, survival, migration, and invasion. Global RNA-sequencing studies demonstrated that PELP1 knockdown significantly reduced the expression of genes involved in the Wnt/ß-catenin pathway. Mechanistic studies demonstrated that PELP1 interacts with and functions as a coactivator of ß-catenin. Knockdown of PELP1 resulted in a significant increase in survival of mice implanted with U87 and GBM PDX models. CONCLUSIONS: PELP1 expression is upregulated in GBM and PELP1 signaling via ß-catenin axis contributes to GBM progression. Thus, PELP1 could be a potential target for the development of therapeutic intervention in GBM.

10.
Oncotarget ; 8(30): 50002-50014, 2017 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-28654894

RESUMEN

Ovarian cancer is the deadliest of all gynecologic cancers. Despite success with initial chemotherapy, the majority of patients relapse with an incurable disease. Development of chemotherapy resistance is a major factor for poor long-term survival in ovarian cancer. The biological effects of estrogens are mediated by estrogen receptor alpha (ERα) and estrogen receptor beta (ERß). Emerging evidence suggests that ovarian cancer cells express ERß that functions as a tumor suppressor; however, the clinical utility of ERß agonists in ovarian cancer remains elusive. We tested the utility of two natural ERß agonists liquiritigenin (Liq), which is isolated from Glycyrrhiza uralensis and S-equol, which is isolated from soy isoflavone daidzein, for treating ovarian cancer. Both natural ERß ligands had significant growth inhibition in cell viability and survival assays, reduced migration and invasion, and promoted apoptosis. Further, ERß agonists showed tumor suppressive functions in therapy-resistant ovarian cancer model cells and sensitized ovarian cancer cells to cisplatin and paclitaxel treatment. Global RNA-Seq analysis revealed that ERß agonists modulate several tumor suppressive pathways, including downregulation of the NF-κB pathway. Immunoprecipitation assays revealed that ERß interacts with p65 subunit of NF-κB and ERß overexpression reduced the expression of NF-κB target genes. In xenograft assays, ERß agonists reduced tumor growth and promoted apoptosis. Collectively, our findings demonstrated that natural ERß agonists have the potential to significantly inhibit ovarian cancer cell growth by anti-inflammatory and pro-apoptotic actions, and natural ERß agonists represent novel therapeutic agents for the management of ovarian cancer.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Productos Biológicos/farmacología , Receptor beta de Estrógeno/agonistas , Estrógenos/farmacología , Neoplasias Ováricas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , FN-kappa B/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Paclitaxel/farmacología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Free Radic Biol Med ; 91: 281-92, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26721594

RESUMEN

The caseinolytic peptidase P (ClpP) is the endopeptidase component of the mitochondrial matrix ATP-dependent ClpXP protease. ClpP degrades unfolded proteins to maintain mitochondrial protein homeostasis and is involved in the initiation of the mitochondrial unfolded protein response (UPR(mt)). Outside of an integral role in the UPR(mt), the cellular function of ClpP is not well characterized in mammalian cells. To investigate the role of ClpP in mitochondrial function, we generated C2C12 muscle cells that are deficient in ClpP using siRNA or stable knockdown using lentiviral transduction. Reduction of ClpP levels by ~70% in C2C12 muscle cells resulted in a number of mitochondrial alterations including reduced mitochondrial respiration and reduced oxygen consumption rate in response to electron transport chain (ETC) complex I and II substrates. The reduction in ClpP altered mitochondrial morphology, changed the expression level of mitochondrial fission protein Drp1 and blunted UPR(mt) induction. In addition, ClpP deficient cells showed increased generation of reactive oxygen species (ROS) and decreased membrane potential. At the cellular level, reduction of ClpP impaired myoblast differentiation, cell proliferation and elevated phosphorylation of eukaryotic initiation factor 2 alpha (eIF2α) suggesting an inhibition of translation. Our study is the first to define the effects of ClpP deficiency on mitochondrial function in muscle cells in vitro. In addition, we have uncovered novel effects of ClpP on mitochondrial morphology, cell proliferation and protein translation pathways in muscle cells.


Asunto(s)
Proliferación Celular , Endopeptidasa Clp/metabolismo , Mitocondrias Musculares/enzimología , Mioblastos/fisiología , Animales , Diferenciación Celular , Línea Celular , Forma de la Célula , Regulación hacia Abajo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Glucólisis , Peróxido de Hidrógeno/metabolismo , Potencial de la Membrana Mitocondrial , Ratones , Mitocondrias Musculares/ultraestructura , Mioblastos/ultraestructura , Biosíntesis de Proteínas , Respuesta de Proteína Desplegada
12.
Cancer Res ; 75(9): 1846-58, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25877877

RESUMEN

The protein tyrosine phosphatase receptor PTPRN2 is expressed predominantly in endocrine and neuronal cells, where it functions in exocytosis. We found that its immature isoform proPTPRN2 is overexpressed in various cancers, including breast cancer. High proPTPRN2 expression was associated strongly with lymph node-positive breast cancer and poor clinical outcome. Loss of proPTPRN2 in breast cancer cells promoted apoptosis and blocked tumor formation in mice, whereas enforced expression of proPTPRN2 in nontransformed human mammary epithelial cells exerted a converse effect. Mechanistic investigations suggested that ProPTPRN2 elicited these effects through direct interaction with TRAF2, a hub scaffold protein for multiple kinase cascades, including ones that activate NF-κB. Overall, our results suggest PTPRN2 as a novel candidate biomarker and therapeutic target in breast cancer.


Asunto(s)
Apoptosis/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/biosíntesis , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Células HCT116 , Células HEK293 , Células HeLa , Xenoinjertos , Humanos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Metástasis Linfática , Células MCF-7 , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , FN-kappa B/metabolismo , Neoplasias/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/metabolismo
13.
Oncotarget ; 4(1): 18-28, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23248157

RESUMEN

Glioma development is a multistep process, involving alterations in genetic and epigenetic mechanisms. Understanding the mechanisms and enzymes that promote epigenetic changes in gliomas are urgently needed to identify novel therapeutic targets. We examined the role of histone demethylase KDM1 in glioma progression. KDM1 was overexpressed in gliomas and its expression positively correlated with histological malignancy. Knockdown of KDM1 expression or its pharmacological inhibition using pargyline or NCL-1 significantly reduced the proliferation of glioma cells. Inhibition of KDM1 promoted up regulation of the p53 target genes p21 and PUMA. Patient-derived primary GBM cells expressed high levels of KDM1 and pharmacological inhibition of KDM1 decreased their proliferation. Further, KDM1 inhibition reduced the expression of stemness markers CD133 and nestin in GBM cells. Mouse xenograft assays revealed that inhibition of KDM1 significantly reduced glioma xenograft tumor growth. Inhibition of KDM1 increased levels of H3K4-me2 and H3K9-Ac histone modifications, reduced H3K9-me2 modification and promoted expression of p53 target genes (p21 and PUMA), leading to apoptosis of glioma xenograft tumors. Our results suggest that KDM1 is overexpressed in gliomas and could be a potential therapeutic target for the treatment of gliomas.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Glioma/tratamiento farmacológico , Histona Demetilasas/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Benzamidas/farmacología , Benzamidas/uso terapéutico , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclopropanos/farmacología , Ciclopropanos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/metabolismo , Glioma/patología , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Humanos , Inmunohistoquímica , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pargilina/farmacología , Pargilina/uso terapéutico , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
Mol Cancer Ther ; 11(5): 1174-82, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22442308

RESUMEN

Gliomas are the most common and devastating central nervous system neoplasms. A gender bias exists in their development: females are at lower risk than males, implicating estrogen-mediated protective effects. Estrogen functions are mediated by two estrogen receptor (ER) subtypes: ERα, which functions as tumor promoter, and ERß, which functions as tumor suppressor. We examined the potential use of ERß agonists as a novel therapeutic to curb the growth of gliomas. Western analysis of six glioma model cells showed detectable expression of ERß with little or no ERα. Treatment of glioma cells with ERß agonists resulted in significant decrease in proliferation. Immunohistochemical analysis of tumor tissues revealed that ERß expression is downregulated in high-grade gliomas. We found that ERß agonists promote both expression and tumor-suppressive functions of ERß in glioma cells. Liquiritigenin, a plant-derived ERß agonist significantly reduced in vivo tumor growth in a xenograft model. Compared with control mice, animals treated with liquiritigenin had greater than 50% reduction in tumor volume and size. Immunohistochemical analysis of tumors revealed a significant increase in the nuclear ERß expression with a concomitant decrease in cell proliferation in the liquiritigenin-treated group. Our results suggest that ERß signaling has a tumor-suppressive function in gliomas. Because ERß agonists are currently in clinical trials and are well tolerated with fewer side effects, identification of an ERß agonist as a therapeutic agent can be readily extended to clinical use with current chemotherapies, providing an additional tool for enhancing survival in glioma patients.


Asunto(s)
Antineoplásicos/farmacología , Receptor beta de Estrógeno/agonistas , Glioma/tratamiento farmacológico , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Receptor beta de Estrógeno/genética , Femenino , Flavanonas/farmacología , Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/patología , Humanos , Ratones , Ratones Desnudos , Transporte de Proteínas , Transducción de Señal
15.
Front Biosci (Schol Ed) ; 3(2): 788-96, 2011 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-21196412

RESUMEN

Integrin-linked kinase 1 (ILK1) is a serine/threonine kinase that plays important roles in a variety of cellular functions including cell survival, migration and angiogenesis. ILK1 is normally expressed in numerous tissues and activated by growth factors, cytokines and hormones. Dysregulation of ILK1 expression or function is found in several hormonal tumors including breast, ovary and prostate. Emerging evidence suggests that ILK overexpression promotes cellular transformation, cell survival, epithelial mesenchymal transition (EMT), and metastasis of hormonal cancer cells while inhibition of ILK1 reduces tumor growth and progression. The recent development of ILK1 inhibitors has provided novel mechanisms for blocking ILK1 signaling to curb metastasis and therapy resistance of hormonal tumors. This review will focus on recent advances made towards understanding the role of ILK signaling axis in progression of hormonal cancer.


Asunto(s)
Neoplasias Hormono-Dependientes/patología , Proteínas Serina-Treonina Quinasas/fisiología , Progresión de la Enfermedad , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/patología , Humanos , Metástasis de la Neoplasia , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/enzimología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal
16.
PLoS One ; 6(6): e21095, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21695158

RESUMEN

BACKGROUND: Proline-, glutamic acid-, and leucine-rich protein (PELP1) is a novel nuclear receptor coregulator with a multitude of functions. PELP1 serves as a scaffolding protein that couples various signaling complexes with nuclear receptors and participates as a transcriptional coregulator. Recent data suggest that PELP1 expression is deregulated in hormonal cancers, and that PELP1 functions as a proto-oncogene; however, the mechanism by which PELP1 promotes oncogenesis remains elusive. METHODOLOGY/PRINCIPAL FINDINGS: Using pharmacological inhibitors, confocal microscopy and biochemical assays, we demonstrated that PELP1 is localized in the nucleolus and that PELP1 is associated with the active ribosomal RNA transcription. Cell synchronization studies showed that PELP1 nucleolar localization varies and the greatest amount of nucleolar localization was observed during S and G2 phases. Using pharmacological compounds and CDK site mutants of PELP1, we found that CDK's activity plays an important role on PELP1 nucleolar localization. Depletion of PELP1 by siRNA decreased the expression of pre-rRNA. Reporter gene assays using ribosomal DNA (pHrD) luc-reporter revealed that PELP1WT but not PELP1MT enhanced the expression of reporter. Deletion of nucleolar domains abolished PELP1-mediated activation of the pHrD reporter. ChIP analysis revealed that PELP1 is recruited to the promoter regions of rDNA and is needed for optimal transcription of ribosomal RNA. CONCLUSIONS/SIGNIFICANCE: Collectively, our results suggest that proto-oncogene PELP1 plays a vital role in rDNA transcription. PELP1 modulation of rRNA transcription, a key step in ribosomal biogenesis may have implications in PELP1-mediated oncogenic functions.


Asunto(s)
Proteínas Co-Represoras/metabolismo , ADN Ribosómico/genética , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Transporte Activo de Núcleo Celular , Ciclo Celular , Nucléolo Celular/metabolismo , Proteínas Co-Represoras/química , Quinasas Ciclina-Dependientes/metabolismo , Células HEK293 , Células HeLa , Humanos , Fosforilación , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/química , Factores de Transcripción/química
17.
Cancer Res ; 70(18): 7166-75, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20807815

RESUMEN

Estrogen receptor (ER) signaling plays an important role in breast cancer progression, and ER functions are influenced by coregulatory proteins. PELP1 (proline-, glutamic acid-, and leucine-rich protein 1) is a nuclear receptor coregulator that plays an important role in ER signaling. Its expression is deregulated in hormonal cancers. We identified PELP1 as a novel cyclin-dependent kinase (CDK) substrate. Using site-directed mutagenesis and in vitro kinase assays, we identified Ser(477) and Ser(991) of PELP1 as CDK phosphorylation sites. Using the PELP1 Ser(991) phospho-specific antibody, we show that PELP1 is hyperphosphorylated during cell cycle progression. Model cells stably expressing the PELP1 mutant that lack CDK sites had defects in estradiol (E2)-mediated cell cycle progression and significantly affected PELP1-mediated oncogenic functions in vivo. Mechanistic studies showed that PELP1 modulates transcription factor E2F1 transactivation functions, that PELP1 is recruited to pRb/E2F target genes, and that PELP1 facilitates ER signaling cross talk with cell cycle machinery. We conclude that PELP1 is a novel substrate of interphase CDKs and that its phosphorylation is important for the proper function of PELP1 in modulating hormone-driven cell cycle progression and also for optimal E2F transactivation function. Because the expression of both PELP1 and CDKs is deregulated in breast tumors, CDK-PELP1 interactions will have implications in breast cancer progression.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Transactivadores/metabolismo , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/fisiología , Línea Celular Tumoral , Proteínas Co-Represoras , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/genética , Factores de Transcripción E2F/genética , Factores de Transcripción E2F/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Mutación , Células 3T3 NIH , Fosforilación , Transactivadores/genética , Factores de Transcripción
18.
J Steroid Biochem Mol Biol ; 118(4-5): 211-8, 2010 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-19800002

RESUMEN

Estradiol (E2), estrogen receptor (ER), ER-coregulators have been implicated in the development and progression of breast cancer. In situ E2 synthesis is implicated in tumor cell proliferation through autocrine or paracrine mechanisms, especially in post-menopausal women. Several recent studies demonstrated activity of aromatase P450 (Cyp19), a key enzyme that plays critical role in E2 synthesis in breast tumors. The mechanism by which tumors enhance aromatase expression is not completely understood. Recent studies from our laboratory suggested that PELP1 (Proline, Glutamic acid, Leucine rich Protein 1), a novel ER-coregulator, functions as a potential proto-oncogene and promotes tumor growth in nude mice models without exogenous E2 supplementation. In this study, we found that PELP1 deregulation contributes to increased expression of aromatase, local E2 synthesis and PELP1 cooperates with growth factor signaling components in the activation of aromatase. PELP1 deregulation uniquely up-regulated aromatase expression via activation of aromatase promoter I.3/II. Analysis of PELP1 driven mammary tumors in xenograft as well as in transgenic mouse models revealed increased aromatase expression. PELP1-mediated induction of aromatase requires functional Src and PI3K pathways. Chromatin immuno precipitation (ChIP) assays revealed that PELP1 is recruited to the Aro 1.3/II aromatase promoter. HER2 signaling enhances PELP1 recruitment to the aromatase promoter and PELP1 plays a critical role in HER2-mediated induction of aromatase expression. Mechanistic studies revealed that PELP1 interactions with orphan receptor ERRalpha, and histone demethylases play a role in the activation of aromatase promoter. Accordingly, ChIP analysis showed alterations in histone modifications at the aromatase promoter in the model cells that exhibit local E2 synthesis. Immunohistochemical analysis of breast tumor progression tissue arrays suggested that deregulation of aromatase expression occurs in advanced-stage and node-positive tumors, and that cooverexpression of PELP1 and aromatase occur in a sub set of tumors. Collectively, our results suggest that PELP1 regulation of aromatase represent a novel mechanism for in situ estrogen synthesis leading to tumor proliferation by autocrine loop and open a new avenue for ablating local aromatase activity in breast tumors.


Asunto(s)
Aromatasa/genética , Neoplasias de la Mama/enzimología , Estrógenos/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Transactivadores/metabolismo , Animales , Aromatasa/metabolismo , Neoplasias de la Mama/genética , Proteínas Co-Represoras , Endometriosis/enzimología , Endometriosis/genética , Epigénesis Genética , Femenino , Humanos , Ratones , Ratones Transgénicos , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Proto-Oncogenes Mas , Receptores de Estrógenos/metabolismo , Transactivadores/genética , Factores de Transcripción , Receptor Relacionado con Estrógeno ERRalfa
19.
Cancer Res ; 70(10): 4092-101, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20460518

RESUMEN

The molecular basis of breast cancer progression to metastasis and the role of estrogen receptor (ER) signaling in this process remain poorly understood. Emerging evidence suggests that ER participates in extranuclear signaling in addition to genomic functions. Recent studies identified proline-, glutamic acid-, and leucine-rich protein-1 (PELP1) as one of the components of ER signalosome in the cytoplasm. PELP1 expression is deregulated in metastatic breast tumors. We examined the mechanism and significance of ER-PELP1-mediated extranuclear signals in the cytoskeletal remodeling and metastasis. Using estrogen dendrimer conjugate (EDC) that uniquely activate ER extranuclear signaling and by using model cells that stably express PELP1 short hairpin RNA (shRNA), we show that PELP1 is required for optimal activation of ER extranuclear actions. Using a yeast two-hybrid screen, we identified integrin-linked kinase 1 (ILK1) as a novel PELP1-binding protein. Activation of extranuclear signaling by EDC uniquely enhanced E2-mediated ruffles and filopodia-like structures. Using dominant-negative and dominant-active reagents, we found that estrogen-mediated extranuclear signaling promotes cytoskeleton reorganization through the ER-Src-PELP1-phosphoinositide 3-kinase-ILK1 pathway. Using in vitro Boyden chamber assays and in vivo xenograft assays, we found that ER extranuclear actions contribute to cell migration. Collectively, our results suggest that ER extranuclear actions play a role in cell motility/metastasis, establishing for the first time that endogenous PELP1 serves as a critical component of ER extranuclear actions leading to cell motility/invasion and that the ER-Src-PELP1-ILK1 pathway represents a novel therapeutic target for preventing the emergence of ER-positive metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Núcleo Celular/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Receptores de Estrógenos/metabolismo , Transactivadores/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/metabolismo , Adhesión Celular , Línea Celular Tumoral , Núcleo Celular/patología , Proliferación Celular , Proteínas Co-Represoras , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Invasividad Neoplásica , Ovariectomía , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Estrógenos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Factores de Transcripción , Familia-src Quinasas/metabolismo
20.
Anal Quant Cytol Histol ; 28(5): 292-6, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17067011

RESUMEN

OBJECTIVE: To study alteration in karyometric variables to distinguish benign from malignant canine mammary tumors. STUDY DESIGN: Formalin-fixed, paraffin-embedded tissue sections from 16 cases of canine mammary tumors were stained with hematoxylin-eosin and analyzed for mean nuclear area, mean nuclear perimeter and shape factor. The morphometric descriptors were then correlated with conventional histologic diagnosis. RESULTS: On Mann-Whitney U-test, nuclear variables, that is, mean nuclear area and mean nuclear perimeter, revealed significant difference between malignant and benign groups (p < 0.01), while shape factor showed significant difference at p < 0.05. All morphometric descriptors showed significant correlation with each other and with histopathologic diagnosis. Contrary to expectations, shape factor was lower in benign compared to malignant tumors. In benign mammary tumors, the highest mean nuclear area and perimeter were 39.59 +/- 10.34 microm2 and 25.39 +/- 4.14 microm, respectively, for fibroadenoma; and in malignant tumors the highest mean nuclear area and perimeter were 48.44 +/- 12.89 microm2 and 27.36 +/- 4.31 microm, respectively, for papillary adenocarcinoma. CONCLUSION: This study provides insight into the alterations in karyometric variables in canine mammary tumors. Mean nuclear area and perimeter appear to have potential to differentiate benign and malignant canine mammary tumors.


Asunto(s)
Enfermedades de los Perros/patología , Cariometría/métodos , Neoplasias Mamarias Animales/patología , Animales , Perros , Femenino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA