Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 316(4): E590-E604, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30668150

RESUMEN

Parathyroid hormone (PTH) and its related peptide (PTH-related peptide 1-34) are two of the Food and Drug Administration-approved bone-promoting drugs for age-related osteoporosis. Treatment with PTH stimulates bone formation. However, the molecular mechanisms of PTH-mediated osteoblast differentiation and cell proliferation are still not completely understood. In this study, we showed that PTH induced endoplasmic reticulum (ER) stress in osteoblasts through the PKR-like endoplasmic reticulum kinase (PERK)-eukaryotic initiation factor 2α (EIF2α)-activating transcription factor 4 (ATF4)-signaling pathway. After separately blocking PERK-EIF2α-ATF4 signaling with two different inhibitors [AMG'44 and integrated stress response inhibitor (ISRIB)] or specific small interfering RNA for PERK and ATF4, the following targets were all downregulated: expression of osteoblast differentiation markers [runt-related transcription factor 2 (Runx2), alkaline phosphatase (Alp), type I collagen (Col1a1), and osteocalcin (Ocn)], cell proliferation markers (CyclinE, CyclinD, and CDC2), amino acid import (Glyt1), and metabolism-related genes (Asns). Additionally, Alp-positive staining cells, Alp activity, matrix mineralization, Ocn secretion, and cell proliferation indexes were inhibited. Interestingly, we found that salubrinal enhanced PTH-induced osteoblast differentiation and proliferation by maintenance of phosphorylation of EIF2α. Furthermore, we observed that PTH increased the association between heat shock protein 90 (HSP90) and PERK and maintained PERK protein stabilization in the early stages of PTH-induced ER stress. Treatment of MC3T3-E1 cells with geldanamycin, an HSP90 inhibitor, decreased PERK protein expression and inhibited osteoblast differentiation and cell proliferation upon PTH treatment. Taken together, our data demonstrate that PTH regulates osteoblast differentiation and cell proliferation, partly by activating the HSP90-dependent PERK-EIF2α-ATF4 signaling pathway.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Hormona Paratiroidea/farmacología , Factor de Transcripción Activador 4/metabolismo , Fosfatasa Alcalina/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Animales , Benzoquinonas/farmacología , Proteína Quinasa CDC2/efectos de los fármacos , Proteína Quinasa CDC2/metabolismo , Línea Celular , Colágeno Tipo I/efectos de los fármacos , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Subunidad alfa 1 del Factor de Unión al Sitio Principal/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Ciclina D/efectos de los fármacos , Ciclina D/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Inhibidores Enzimáticos/farmacología , Factor 2 Eucariótico de Iniciación/metabolismo , Proteínas de Transporte de Glicina en la Membrana Plasmática/efectos de los fármacos , Proteínas de Transporte de Glicina en la Membrana Plasmática/metabolismo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/metabolismo , Lactamas Macrocíclicas/farmacología , Ratones , Osteoblastos/metabolismo , Osteocalcina/efectos de los fármacos , Osteocalcina/metabolismo , Transducción de Señal , eIF-2 Quinasa/metabolismo
2.
Asian J Androl ; 17(6): 996-1005, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26289399

RESUMEN

This study was designed to explore the regulatory effects of male germ cell secreting factor NODAL on Sertoli cell fate decisions from obstructive azoospermia (OA) and nonobstructive azoospermia (NOA) patients. Human Sertoli cells and male germ cells were isolated using two-step enzymatic digestion and SATPUT from testes of azoospermia patients. Expression of NODAL and its multiple receptors in human Sertoli cells and male germ cells were characterized by reverse transcription-polymerase chain reaction (RT-PCR) and immunochemistry. Human recombinant NODAL and its receptor inhibitor SB431542 were employed to probe their effect on the proliferation of Sertoli cells using the CCK-8 assay. Quantitative PCR and Western blots were utilized to assess the expression of Sertoli cell functional genes and proteins. NODAL was found to be expressed in male germ cells but not in Sertoli cells, whereas its receptors ALK4, ALK7, and ACTR-IIB were detected in Sertoli cells and germ cells, suggesting that NODAL plays a regulatory role in Sertoli cells and germ cells via a paracrine and autocrine pathway, respectively. Human recombinant NODAL could promote the proliferation of human Sertoli cells. The expression of cell cycle regulators, including CYCLIN A, CYCLIN D1 and CYCLIN E, was not remarkably affected by NODAL signaling. NODAL enhanced the expression of essential growth factors, including GDNF, SCF, and BMP4, whereas SB431542 decreased their levels. There was not homogeneity of genes changes by NODAL treatment in Sertoli cells from OA and Sertoli cell-only syndrome (SCO) patients. Collectively, this study demonstrates that NODAL produced by human male germ cells regulates proliferation and numerous gene expression of Sertoli cells.


Asunto(s)
Azoospermia/metabolismo , Proliferación Celular/efectos de los fármacos , Proteína Nodal/farmacología , Proteínas Recombinantes/farmacología , Células de Sertoli/efectos de los fármacos , Espermatozoides/metabolismo , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo II/metabolismo , Adulto , Comunicación Autocrina , Benzamidas/farmacología , Western Blotting , Proteína Morfogenética Ósea 4/efectos de los fármacos , Proteína Morfogenética Ósea 4/metabolismo , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Dioxoles/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Inmunohistoquímica , Masculino , Proteína Nodal/metabolismo , Comunicación Paracrina , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Células Madre/efectos de los fármacos , Factor de Células Madre/metabolismo
3.
Womens Health (Lond) ; 10(1): 45-57, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24328598

RESUMEN

Uterine serous carcinoma (USC) is a highly aggressive variant of endometrial cancer. Although it only represents less than 10% of all cases, it accounts for a disproportionate number of deaths from endometrial cancer. Comprehensive surgical staging followed by carboplatin and paclitaxel chemotherapy represents the mainstay of USC therapy. Vaginal cuff brachytherapy is also of potential benefit in USC. Recent whole-exome sequencing studies have demonstrated gain of function of the HER2/NEU gene, as well as driver mutations in the PIK3CA/AKT/mTOR and cyclin E/FBXW7 oncogenic pathways in a large number of USCs. These results emphasize the relevance of these novel therapeutic targets for biologic therapy of chemotherapy-resistant recurrent USC.


Asunto(s)
Antineoplásicos/uso terapéutico , Cistadenocarcinoma Seroso/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Uterinas/tratamiento farmacológico , Fosfatidilinositol 3-Quinasa Clase I , Ciclina E/efectos de los fármacos , Ciclina E/genética , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Genes erbB-2/efectos de los fármacos , Genes erbB-2/genética , Humanos , Mutación , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Ensayos Clínicos Controlados Aleatorios como Asunto , Neoplasias Uterinas/genética , Neoplasias Uterinas/patología
4.
Cell Cycle ; 11(7): 1374-82, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22421144

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) is the sixth leading cancer in the world; the main risk factors are alcohol and tobacco use. Advancements in therapies have yet to improve the prognosis of HNSCC. The connection between diabetes and cancer is being recognized, and metformin has been shown to decrease cancer incidence in diabetic patients. Accordingly, here, for the first time, we investigated metformin's efficacy on the growth and viability of human HNSCC FaDU and Detroit cells. Our results show that metformin treatment (5-20 mM) dose-dependently inhibits the growth of both cell lines. In FaDU cells, metformin caused 18-57% and 35-81% growth inhibition after 48 and 72 h treatments, respectively. Similarly, in Detroit 562 cells, 48 and 72 h metformin treatment resulted in 20-57% and 33-82% inhibition, respectively. Mechanistically, metformin caused G 1 arrest, which coincided with a decrease in the protein levels of CDKs (2, 4 and 6), cyclins (D1 and E) and CDK inhibitors (p15, p16, p18 and p27), but no change in p19 and p21. Metformin also decreased the levels of oncogenic proteins Skp2 and ß-Trcp. In other studies, metformin decreased the phosphorylation of 4E-BP1 at Ser65, Thr37/46 and Thr70 sites, but drastically increased the phosphorylation of EF2 at Thr56 and AMPK at Thr172, which results in global translational inhibition. In summary, the observed wide spectrum of mechanistic effects of metformin on HNSCC cells provides support for the anticancer capability of the drug and its potential use in future therapies.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/metabolismo , Metformina/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Ciclina D1/metabolismo , Ciclina E/análisis , Ciclina E/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Proteínas de Neoplasias/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello , Proteínas con Repetición de beta-Transducina/metabolismo , Proteínas de Unión al GTP rho/metabolismo
5.
Anticancer Drugs ; 23(4): 370-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22185819

RESUMEN

Artemisinin, a sesquiterpene phytolactone derived from Artemisia annua, is a potent antimalarial compound with promising anticancer properties, although the mechanism of its anticancer signaling is not well understood. Artemisinin inhibited proliferation and induced a strong G1 cell cycle arrest of cultured MCF7 cells, an estrogen-responsive human breast cancer cell line that represents an early-stage cancer phenotype, and effectively inhibited the in-vivo growth of MCF7 cell-derived tumors from xenografts in athymic nude mice. Artemisinin also induced a growth arrest of tumorigenic human breast cancer cell lines with preneoplastic and late stage cancer phenotypes, but failed to arrest the growth of a nontumorigenic human mammary cell line. Concurrent with the cell cycle arrest of MCF7 cells, artemisinin selectively downregulated the transcript and protein levels of the CDK2 and CDK4 cyclin-dependent kinases, cyclin E, cyclin D1, and the E2F1 transcription factor. Analysis of CDK2 promoter-luciferase reporter constructs showed that the artemisinin ablation of CDK2 gene expression was accounted for by the loss of CDK2 promoter activity. Chromatin immunoprecipitation revealed that artemisinin inhibited E2F1 interactions with the endogenous MCF7 cell CDK2 and cyclin E promoters. Moreover, constitutive expression of exogenous E2F1 prevented the artemisinin-induced cell cycle arrest and downregulation of CDK2 and cyclin E gene expression. Taken together, our results demonstrate that the artemisinin disruption of E2F1 transcription factor expression mediates the cell cycle arrest of human breast cancer cells and represents a critical transcriptional pathway by which artemisinin controls human reproductive cancer cell growth.


Asunto(s)
Antineoplásicos/farmacología , Artemisininas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Factor de Transcripción E2F1/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Factor de Transcripción E2F1/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Exp Ther Oncol ; 9(3): 175-81, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22070048

RESUMEN

Ovarian cancer is the leading cause of death from gynecological cancer. The high mortality rate reflets the lack of early diagnosis and limited treatment alternatives. We have observed a number of properties of zinc cytotoxicity that make it attractive from a therapeutic standpoint. Using SKOV3 and ES2 cells, ovarian cancer cell lines that demonstrate varied degrees of resistance to known therapeutics, we show that zinc killing is time and concentration dependent. Death is preceded by distinct changes in cell shape and size. The effects of zinc are additive with cisplatin or doxorubicin, whose morphological effects are distinct from those of zinc. Cytotoxicity of paclitaxel is minimal, making it difficult to determine additivity with zinc. Paclitaxel results in changes in cell shape and size similar to those of zinc but has different effects on cell cycle progression and cyclin expression. The data indicate that the means by which zinc kills ovarian cancer cells is distinct from currently used chemotherapeutics. Based on the properties reported here, zinc has the potential to be developed as either a primary treatment or as a second line of defense against cancers that have developed resistance to currently used chemotherapeutics.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Oligoelementos/farmacología , Zinc/farmacología , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/farmacología , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina D/efectos de los fármacos , Ciclina D/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Paclitaxel/farmacología , Piridinas/farmacología , Tionas/farmacología
7.
Arch Oral Biol ; 55(12): 938-45, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20832772

RESUMEN

Voltage gated chloride channels (ClCs) play an important role in the regulation of intracellular pH and cell volume homeostasis. Mutations of these genes result in genetic diseases with abnormal bone deformation and body size, indicating that ClCs may have a role in chondrogenesis. In the present study, we isolated chicken mandibular mesenchymal cells (CMMC) from Hamburg-Hamilton (HH) stage 26 chick embryos and induced chondrocyte maturation by using ascorbic acid and ß-glycerophosphate (AA-BGP). We also determined the effect of the chloride channel inhibitor NPPB [5-nitro-2-(3-phenylpropylamino) benzoic acid] on regulation of growth, differentiation, and gene expression in these cells using MTT and real-time PCR assays. We found that CLCN1 and CLCN3-7 mRNA were expressed in CMMC and NPPB reduced expression of CLCN3, CLCN5, and CLCN7 mRNA in these cells. At the same time, NPPB inhibited the growth of the CMMC, but had no effect on the mRNA level of cyclin D1 and cyclin E (P>0.05) with/without AA-BGP treatment. AA-BGP increased markers for early chondrocyte differentiation including type II collagen, aggrecan (P<0.01) and Sox9 (P<0.05), whilst had no effect on the late chondrocyte differentiation marker type X collagen. NPPB antagonized AA-BGP-induced expression of type II collagen and aggrecan (P<0.05). Furthermore, NPPB downregulated type X collagen (P<0.05) with/without AA-BGP treatment. We conclude that abundant chloride channel genes in CMMC play important roles in regulating chondrocyte proliferation and differentiation. Type X collagen might function as a target of chloride channel inhibitors during the differentiation process.


Asunto(s)
Canales de Cloruro/fisiología , Condrogénesis/fisiología , Mandíbula/embriología , Mesodermo/embriología , Agrecanos/efectos de los fármacos , Animales , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Canales de Cloruro/análisis , Canales de Cloruro/antagonistas & inhibidores , Condrocitos/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Colágeno Tipo II/efectos de los fármacos , Colágeno Tipo X/efectos de los fármacos , Ciclina D1/efectos de los fármacos , Ciclina E/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glicerofosfatos/farmacología , Mandíbula/efectos de los fármacos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Proteínas Musculares/análisis , Proteínas Musculares/antagonistas & inhibidores , Nitrobenzoatos/farmacología , Factor de Transcripción SOX9/efectos de los fármacos
8.
Brain Res ; 1359: 272-80, 2010 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-20813099

RESUMEN

Methylmercury (MeHg) is a potent environmental neurotoxin that shows toxicity to developing central nervous system (CNS), causing brain damage in children even at low exposure levels. However, the mechanisms for its effect on CNS are not well understood. In current study, primary cultures of progenitor cells from embryonic cerebral cortex were used as a model system to study the potential effect and the underlying mechanism of MeHg on neural progenitor cells. Results showed that, in cultured cortical progenitor cells, 48-h exposure to low-level of MeHg (at 2.5 nM, 5 nM and 50 nM, respectively) caused G1/S cell cycle arrest in a dose-dependent manner without inducing cell death. Interestingly, the expression of cyclin E, which promotes G1/S transition, but not cyclin D1 and CDK2, was selectively downregulated by exposure of MeHg. In addition, low-level of MeHg inhibited the maintenance of ERK1/2 phosphorylation, possibly by abolishing the late phase ERK1/2 activation induced by bFGF. Thus, MeHg may induce proliferation inhibition and cell cycle arrest of neural progenitor cells via regulating cyclin E expression and perturbing a pathway that involves ERK1/2.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Compuestos de Metilmercurio/toxicidad , Células-Madre Neurales/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Células-Madre Neurales/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Breast Cancer Res Treat ; 121(2): 355-64, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19641990

RESUMEN

In contrast to deoxyribose or arabinose containing nucleoside analogs that are currently established for cancer therapeutics, 8-chloro-adenosine (8-Cl-Ado) possesses a ribose sugar. This unique nucleoside analog is RNA-directed and is in a phase I clinical trial for hematological malignancies. RNA-directed therapies are effective for the treatment of many malignancies as their activities are primarily aimed at short-lived transcripts, which are typically encoded by genes that promote the growth and survival of tumor cells such as cyclin E in breast cancer. Based on this, we hypothesized that 8-Cl-Ado, a transcription inhibitor, will be effective for the treatment of breast cancer cells. The metabolism of 8-Cl-Ado and the effect on ATP in the breast cancer cell lines MCF-7 and BT-474 were measured using HPLC analysis. In these cells, 8-Cl-Ado was effectively taken up, converted to its cytotoxic metabolite, 8-Cl-ATP, and depleted the endogenous ATP levels. This in turn led to an inhibition of RNA synthesis. The RNA synthesis inhibition was associated with a depletion of cyclin E expression, which is indicative of a diminished tumorigenic phenotype. The final outcome of 8-Cl-Ado treatment of the breast cancer cells was growth inhibition due to an induction of apoptosis and a loss of clonogenic survival. These results indicate that 8-Cl-Ado, which is currently in clinic for hematological malignancies, may be an effective agent for the treatment of breast cancer.


Asunto(s)
2-Cloroadenosina/análogos & derivados , Antineoplásicos/metabolismo , Neoplasias de la Mama/metabolismo , Ciclina E/metabolismo , ARN/efectos de los fármacos , 2-Cloroadenosina/metabolismo , 2-Cloroadenosina/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Separación Celular , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Ciclina E/efectos de los fármacos , Femenino , Citometría de Flujo , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Anticancer Drugs ; 20(4): 230-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19288605

RESUMEN

A number of polyamine analogs are currently used in various clinical trials as cancer treatment and it is important to investigate their effects not only on cancer cells but also on normal cells. Treatment with polyamine analogs depletes cells of polyamines and inhibits cell proliferation, but the analogs cannot take over the normal function of the natural polyamines in the cell. In this study, the normal-like breast epithelial cell line MCF-10A was treated with the polyamine analog N',N"-diethylnorspermine (DENSPM). The cells were then studied using a bromodeoxyuridine- DNA flow cytometry method as well as western blot. The ability of both normal-like and breast cancer cells to recover from DENSPM treatment was also studied. DENSPM treatment of MCF-10A cells resulted in a prolongation of the S and G2 +M phases, followed by a G1/S block. The p53/p21/RB1 pathway was involved in the G1/S block as shown by increased levels of p53 and p21 detected by western blot. Decreased levels of cyclin E1, cyclin A2, and cyclin B1 in DENSPM-treated cells can explain the prolongation of cell cycle phases that occurred before the G1/S block. We also show that MCF-10A cells rapidly recover from DENSPM-induced growth inhibition in contrast to four human breast cancer cell lines. The goal of cancer treatment is to cause minimal and reversible damage to normal cells, while cancer cells should be eliminated. Altogether, the data show that treatment with polyamine analogs spares normal cells, while negatively affecting the cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Espermina/análogos & derivados , Western Blotting , Neoplasias de la Mama/patología , Bromodesoxiuridina , Línea Celular , Línea Celular Tumoral , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina A2 , Ciclina B/efectos de los fármacos , Ciclina B/metabolismo , Ciclina B1 , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , ADN , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Citometría de Flujo , Humanos , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Espermina/farmacología
11.
Biochem Pharmacol ; 77(2): 151-8, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18977205

RESUMEN

Cyclin E is the Cdk2-regulatory subunit required for the initiation of DNA replication at the G1/S transition. It accumulates in late G1 phase and gets rapidly degraded by the ubiquitin/proteasome pathway during S phase. The degradation of cyclin E is a consequence of its phosphorylation and subsequent isomerization by the peptidyl-prolyl isomerase Pin1. We show that in the colon cancer cells HT-29 the inhibition of the chaperone function of Hsp90 by geldanamycin (GA) enhances the ubiquitinylation of cyclin E and triggers active degradation via the proteasome pathway. As Hsp90 forms multiprotein complexes with and regulates the function and cell contents of numerous signaling proteins, this observation suggests a direct interaction between Hsp90 and cyclin E. However, experiments using cell lysate fractionation did not reveal the presence of complexes containing both Hsp90 and cyclin E. Coupled transcription/translation experiments also failed to detect the formation of complexes between newly synthesized cyclin E and Hsp90. We conclude that Hsp90 can regulate the degradation of cellular proteins without binding to them, by an indirect mechanism. This conclusion postulates a new category of proteins that are affected by the inactivation of Hsp90. Our observations do not support the possible involvement of a PPIase in this indirect mechanism. Besides, we did not observe active geldanamycin-dependent degradation of cyclin E in the prostate cancer-derived cell line DU-145, indicating that the Hsp90-dependent stabilization of cyclin E requires specific regulatory mechanism which may be lost in certain types of cancer cells.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Benzoquinonas/farmacología , Ciclina E/metabolismo , Proteínas HSP90 de Choque Térmico/fisiología , Lactamas Macrocíclicas/farmacología , Afidicolina/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon , Ciclina E/efectos de los fármacos , Ciclina E/genética , Replicación del ADN/fisiología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Homeostasis , Humanos , Masculino , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/efectos de los fármacos , Neoplasias de la Próstata , Biosíntesis de Proteínas , Transcripción Genética , Ubiquitina/metabolismo
12.
Environ Toxicol ; 24(3): 243-58, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18683188

RESUMEN

Humic acid (HA) in well water used by the inhabitants for drinking is one of the possible etiological factors for Blackfoot disease (BFD). In this study, the ability of HA to inhibit cell cycle progression and induce apoptosis in cultured smooth muscle cells (SMCs; A7r5) was investigated. Treatment of the SMCs at various HA concentrations (25-200 microg/mL) resulted in sequences of events marked by apoptosis, as shown by loss of cell viability, morphology change, and internucleosomal DNA fragmentation. HA-induced apoptotic cell death that is associated with loss of mitochondrial membrane potential (Delta Psi m), cytochrome c translocation, caspase-3, -8, and -9 activation, poly ADP-ribose polymerase (PARP) degradation, dysregulation of Bcl-2 and Bax, and upregulation of p53 and phospholyrated p53 (p-p53) in SMCs. Flow cytometry analysis demonstrated that HA blocked cell cycle progress in the G1 phase in SMCs. This blockade of cell cycle was associated with reduced amounts of cyclin D1, CDK4, cyclin E, CDK2, and hyperphosphorylated retinoblastoma protein (pRb) in a time-dependent manner. Apparent DNA strand breaks (DNA damage) were also detected in a dose-dependent manner using Single-cell gel electrophoresis assay (comet assay). Furthermore, HA induced dose-dependent elevation of reactive oxygen species (ROS) level in SMCs, and antioxidant vitamin C and Trolox effectively suppressed HA-induced DNA damage and dysregulation of Bcl-2/Bax. Our findings suggest that HA-induced DNA damage, cell cycle arrest, and apoptosis in SMCs may be an underlying mechanisms for the atherosclerosis and thrombosis observed in the BFD endemic region.


Asunto(s)
Apoptosis , Fase G1/efectos de los fármacos , Sustancias Húmicas/toxicidad , Músculo Liso Vascular/efectos de los fármacos , Animales , Caspasas/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo XI/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/genética , Ciclina E/efectos de los fármacos , Ciclina E/genética , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/genética , Citocromos c/metabolismo , Fragmentación del ADN , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Proteína de Retinoblastoma/metabolismo , Proteína X Asociada a bcl-2/metabolismo
13.
Virology ; 367(1): 1-9, 2007 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-17568647

RESUMEN

Epidemiological studies have demonstrated that 15 different mucosal human papillomavirus (HPV) types of the genus alpha of the HPV phylogetic tree are classified as high risk for cervical cancer development. Three additional HPV types of the same genus, HPV26, 53 and 66, are classified as probable high-risk types. In this study, we have characterized the biological properties of the E7 oncoproteins from these three HPV types. All of the corresponding E7 proteins were able to associate with retinoblastoma protein (pRb) and up-regulated the expression of several positive cell cycle regulators, i.e. CDK2, cyclin A and cylin E. However, HPV26 E7 appears to be more efficient than HPV53 and 66 E7 in up-regulating the transcription of cyclin A. Unlike E7 from the high-risk type HPV16 protein, HPV26, 53 and 66 did not efficiently promote pRb degradation. In addition, E7 from these viruses was able to promote proliferation of primary human keratinocytes and circumvent G1 arrest imposed by overexpression of p16(INK4a), but with less efficiency than the high-risk HPV16 E7. Together, our data show that in vitro properties of these E7 proteins correlate with the epidemiological classification of HPV26, 53 and 66 as HPV types with an intermediate risk for cervical cancer development.


Asunto(s)
Alphapapillomavirus/clasificación , Alphapapillomavirus/patogenicidad , Transformación Celular Neoplásica , Queratinocitos/virología , Proteínas E7 de Papillomavirus/metabolismo , Alphapapillomavirus/metabolismo , Línea Celular , Línea Celular Transformada , Células Cultivadas , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fase G1/efectos de los fármacos , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/farmacología , Infecciones por Papillomavirus/virología , Proteína de Retinoblastoma/metabolismo , Factores de Riesgo , Regulación hacia Arriba , Neoplasias del Cuello Uterino/virología
14.
Br J Cancer ; 95(11): 1514-24, 2006 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-17088910

RESUMEN

The cisplatin analogue 1R,2R-diaminocyclohexane(trans-diacetato)(dichloro)platinum(IV) (DAP) is a DNA-damaging agent that will be entering clinical trials for its potent cytotoxic effects against cisplatin-resistant tumour cells. This cytotoxicity may reside in its ability to selectively activate G1-phase checkpoint response by inhibiting CDKs via the p53/p21 pathway. We have now evaluated the role of another CDK inhibitor p27 as a contributor to DAP-mediated inhibition of G1-phase CDK2 activity. Our studies in ovarian A2780 tumour cells demonstrate that p27 levels induced by DAP are comparable to or greater than those seen for p21. The induction of p27 is not through a transcriptional mechanism, but rather is due to a four-fold increase in protein stabilisation through a mechanism dependent on p21. Moreover, DAP-induced p21 promoted the selective increase of p27 in the CDK2 complex, but not in CDK4 complex, and this selective increase contributed to inhibition of the CDK2 kinase activity. The inhibited complex contained either p27 or p21, but not both, with the relative levels of cyclin E associated with p27 and p21 indicating that about 25% of the inhibition of CDK2 activity was due to p27 and 75% due to p21. This study provides the first evidence that p27 upregulation is directly attributable to activation of the p53/p21 pathway by a DNA-damaging agent, and promulgates p53/p21/p27 axis as a significant component of checkpoint response.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/análogos & derivados , Ciclina E/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Daño del ADN , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Northern Blotting , Línea Celular Tumoral , Cisplatino/farmacología , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Quinasas Ciclina-Dependientes/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Femenino , Humanos , Immunoblotting , Inmunoprecipitación , Compuestos Organoplatinos , Plásmidos , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Interferente Pequeño , Transfección , Regulación hacia Arriba
15.
Cancer Lett ; 234(2): 199-208, 2006 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-15885897

RESUMEN

Caffeic acid phenethyl ester (CAPE), a component of honeybee propolis, has been reported to hold various biochemical responses. In the preliminary study, we found that CAPE inhibited the growth of C6 glioma cells in a dose dependent and time dependent manner as shown by the results of trypan blue dye exclusion assay and cell proliferation assay. In addition, the cell number percentage of the G0/G1 phase increased to 85% after the treatment with 50 microM of CAPE for 24h. After treatment with CAPE (50 microM) for 6h, it demonstrated that the protein level of hyperphosphorylated pRb decreased, and cyclin dependent kinase inhibitors p21, p27, and p16 were marked up-regulated. The association of CDK2 and cyclin E that affects the CDK2 activity decreased. When C6 cells were grown as xenografts in nude mice, treatment with CAPE (1-10mg/kg; ip) induced a significant dose dependent decrease in tumor growth by evaluating tumor volume and tumor weight. Histochemical and immunohistochemical analysis revealed that CAPE treatment significantly reduced the number of mitotic cells and proliferating cell nuclear antigen (PCNA)-positive cells in C6 glioma. These results suggest that CAPE presents an antitumor potential for glioma by inhibiting the growth of tumor cells.


Asunto(s)
Ácidos Cafeicos/farmacología , Proliferación Celular/efectos de los fármacos , Glioma/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Alcohol Feniletílico/análogos & derivados , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclina E/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Immunoblotting , Inmunohistoquímica , Técnicas In Vitro , Ratones , Alcohol Feniletílico/farmacología , Ratas , Proteína de Retinoblastoma/efectos de los fármacos
16.
Int J Artif Organs ; 28(1): 44-50, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15742309

RESUMEN

In mammalian cells, cellular differentiation into specific cell types is usually preceded by growth arrest. On the other hand, the induced differentiation may also be preceded by an enhanced G1-S transition of the cell cycle prior to the growth arrest. This suggests that an early increase in proliferation is in some way a prerequisite for subsequent differentiation. We therefore attempted to assess whether we could produce human hepatocytes with further differentiated functions by promoting G1-S transition in a butyrate-treated human hepatocyte cell line. A cyclin E-over-expressing cell line was established by transfecting human cyclin E cDNA. Upon butyrate treatment, the cyclin E-over-expressing cells exhibited a significantly increased albumin-secreting and ammonia-detoxifying capacity when compared to the control cells. In particular, the ornithine transcarbamylase activity was increased in these cells. Collectively, these results implicate that the cyclin E over-expression may augment the hepatocyte-specific functions during the butyrate-induced differentiation process of human hepatocytes by enhancing G1-S cell cycle transition.


Asunto(s)
Butiratos/farmacología , Ciclina E/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Albúminas/metabolismo , Amoníaco/antagonistas & inhibidores , Proteínas Potenciadoras de Unión a CCAAT/análisis , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular , Ciclina E/genética , Proteínas de Unión al ADN/análisis , Fase G1/efectos de los fármacos , Regulación de la Expresión Génica/genética , Factor Nuclear 4 del Hepatocito , Humanos , Ornitina Carbamoiltransferasa/efectos de los fármacos , Fosfoproteínas/análisis , Fase S/efectos de los fármacos , Factor de Transcripción CHOP , Factores de Transcripción/análisis , Transfección , Urea/análisis
17.
Anticancer Drugs ; 15(6): 625-32, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15205608

RESUMEN

3-Hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors have been developed as lipid-lowering drugs, and are well recognized to reduce morbidity and mortality from coronary artery disease. Several recent experimental studies have focused on the inhibitory effects of HMG-CoA reductase inhibitor on tumor cell growth in vitro and in vivo, dependent on a direct effect on cancer cells. In the present study, we aimed to investigate the potential anti-angiogenic effect of pravastatin and its mechanism of action. Using human umbilical vein endothelial cells (HUVECs) as a model of angiogenesis, we investigated the effect of pravastatin on the various steps of angiogenesis, including endothelial cell proliferation and adhesion to extracellular matrix proteins. Pravastatin induced a dose-dependent decrease in the proliferative activity of endothelial cells, which was dependent on the cell cycle arrest to the G1 phase and not on cell apoptosis. G1 arrest was due to the decrease of cyclin D, cyclin E and cyclin-dependent kinase 2 levels. In addition, pravastatin inhibited tube formation on Matrigel and adhesion to extracellular matrix, but did not affect matrix metalloproteinase production. The present results demonstrate the anti-angiogenic activity of pravastatin and its potential use as an anticancer drug is suggested.


Asunto(s)
Endotelio Vascular/citología , Fase G1/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Neovascularización Patológica/prevención & control , Pravastatina/farmacología , Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Quinasas CDC2-CDC28/efectos de los fármacos , Quinasas CDC2-CDC28/genética , Quinasas CDC2-CDC28/metabolismo , División Celular/efectos de los fármacos , Células Cultivadas , Ciclina D , Ciclina E/efectos de los fármacos , Ciclina E/genética , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina , Ciclinas/efectos de los fármacos , Ciclinas/genética , Ciclinas/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Metaloproteinasa 2 de la Matriz/química , Metaloproteinasa 9 de la Matriz/química , Neovascularización Patológica/tratamiento farmacológico , Fosfatos de Poliisoprenilo/farmacología , Pravastatina/antagonistas & inhibidores , Pravastatina/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Sesquiterpenos , Venas Umbilicales/citología , Venas Umbilicales/efectos de los fármacos
18.
J Oral Sci ; 46(1): 37-44, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15141722

RESUMEN

It has previously been demonstrated that gingival fibroblasts derived from nifedipine-reactive patients (nifedipine responders) show a greater cell proliferation rate than those from nifedipine non-reactive patients (nifedipine non-responders) in the presence of 1 microM nifedipine. The aim of the present study was to characterize cell cycle differences between nifedipine responder and non-responder fibroblast cells and determine the effect of basic fibroblast growth factor (bFGF) on cell cycle progression. Further, the effect of bFGF on cyclins A, B1, D1, E, and CDKs 1, 2, 4, 6 mRNA expression in responder and non-responder cells was investigated. A population of nifedipine responder cells underwent progression to S and G2/M phases from G0/G1 phase in the presence of 10% fetal calf serum or 10 ng/ml bFGF was greater than nifedipine non-responder cells. mRNA expression of cyclins A, B1, D1, E and CDKs 1, 2, 4, 6 in the presence of 10 ng/ml bFGF was generally greater in nifedipine responder cells than non-responder cells. These results indicate that nifedipine responder cells may be more susceptible to growth factors such as bFGF with a resultant increase in expression of cyclins and CDKs in responder compared with non-responder cells.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/efectos de los fármacos , Encía/efectos de los fármacos , Sobrecrecimiento Gingival/patología , Nifedipino/efectos adversos , Proteínas Proto-Oncogénicas , Proteína Quinasa CDC2/efectos de los fármacos , Quinasas CDC2-CDC28/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Ciclina A/efectos de los fármacos , Ciclina B/efectos de los fármacos , Ciclina B1 , Ciclina D1/efectos de los fármacos , Ciclina E/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/efectos de los fármacos , Fase G1/efectos de los fármacos , Fase G2/efectos de los fármacos , Encía/citología , Sobrecrecimiento Gingival/fisiopatología , Humanos , Mitosis/efectos de los fármacos , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Fase S/efectos de los fármacos
19.
Int J Cancer ; 109(5): 643-52, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-14999769

RESUMEN

The effects of GA, an ansamycin antibiotic in development as a lead anticancer drug, were studied in mouse BP-A31 fibroblasts and in human cancer-derived cell lines. GA and related molecules act by inhibiting the chaperone function of the Hsp90 protein through competition for ATP binding. The antiproliferative effects of GA have been attributed to destabilization of the Raf-1 protein, one of the targets of Hsp90, and to the resulting inhibition of MAPK. Addition of GA to BP-A31 cells, synchronously progressing through the G(1) phase, inhibited Rb hyperphosphorylation and G(1)/S transition irrespective of the time of addition. The G(1) arrest was accompanied by a progressive decrease in Raf-1 content, especially of the phosphorylated form; however, GA caused only partial inhibition of MAPK phosphorylation. We show that GA triggers a rapid and marked decrease in the kinase activity of the cyclin E/cdk2 complex coupled with a decline in both total and cdk2-associated cyclin E. In transient transfection experiments, inhibition of cyclin E expression by GA was correlated with inhibition of the transcriptional activity of the cyclin E gene promoter. Inhibition of cdk4 activity by GA was observed 3 hr after addition of the drug to late G(1) cells but not after a short (1 hr) exposure, as revealed by the phosphorylation of Rb on the Ser(780) residue. In human cancer-derived cell lines expressing or not a functional Rb protein, GA blocked proliferation and inhibited the transcriptional activity of the cyclin E gene promoter. In these cell lines, the antiproliferative effect of GA was not limited to the G(1) phase, suggesting the existence of multiple cellular targets of the drug.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Quinasas CDC2-CDC28/efectos de los fármacos , Ciclina E/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Interfase/efectos de los fármacos , Quinonas/farmacología , Benzoquinonas , Western Blotting , Quinasas CDC2-CDC28/metabolismo , Ciclo Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lactamas Macrocíclicas , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Chaperonas Moleculares/antagonistas & inhibidores , Fosforilación/efectos de los fármacos , Plásmidos/metabolismo , Pruebas de Precipitina , Regiones Promotoras Genéticas/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteína de Retinoblastoma/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Transfección , Células Tumorales Cultivadas
20.
Am J Pathol ; 164(3): 937-46, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14982847

RESUMEN

Lipoxins (LX) are endogenously produced eicosanoids with a spectrum of bioactions that suggest anti-inflammatory, pro-resolution roles for these agents. Mesangial cell (MC) proliferation plays a pivotal role in the pathophysiology of glomerular inflammation and is coupled to sclerosis and tubulointerstitial fibrosis. We have previously reported that LXA4 acts through a specific G-protein-coupled-receptor (GPCR) to modulate MC proliferation in response to the proinflammatory mediators LTD4 and platelet-derived growth factor (PDGF). Further investigations revealed that these effects were mediated by modulation of receptor tyrosine kinase activity. Here we have explored the underlying mechanisms and report inhibition of growth factor (PDGF; epithelial growth factor) activation of Akt/PKB by LXA4. LXA4 (10 nmol/L) modulates PDGF-induced (10 ng/ml, 24 hours) decrements in the levels of cyclin kinase inhibitors p21Cip1 and p27Kip1. PDGF-induced increases in CDK2-cyclin E complex formation are also inhibited by LXA4. The potential of LXA4 as an anti-inflammatory therapeutic is compromised by its degradation; this has been circumvented by synthesis of stable analogs. We report that 15-(R/S)-methyl-LXA4 and 16-phenoxy-LXA4 mimic the native compound with respect to modulation of cell proliferation and PDGF-induced changes in cell cycle proteins. In vivo, MC proliferation in response to PDGF is associated with TGFbeta1 production and the subsequent development of renal fibrosis. Here we demonstrate that prolonged (24 to 48 hours) exposure to PDGF is associated with autocrine TGFbeta1 production, which is significantly reduced by LXA4. In aggregate these data demonstrate that LX inhibit PDGF stimulated proliferation via modulation of the PI-3-kinase pathway preventing mitogen-elicited G1-S phase progression and suggest the therapeutic potential of LX as anti-fibrotic agents.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Mesangio Glomerular/efectos de los fármacos , Sustancias de Crecimiento/metabolismo , Lipoxinas/farmacología , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/efectos de los fármacos , Western Blotting , Quinasas CDC2-CDC28/efectos de los fármacos , Quinasas CDC2-CDC28/metabolismo , Proteínas de Ciclo Celular/efectos de los fármacos , Células Cultivadas , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Mesangio Glomerular/citología , Humanos , Inmunohistoquímica , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-akt , Proteínas Supresoras de Tumor/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...