Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Oncogene ; 35(32): 4165-78, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-26686086

RESUMEN

Mammary stem cells (MSCs) expansion is associated with aggressive human breast cancer. The nuclear receptor peroxisome proliferator activated receptor γ (PPARγ) is a breast cancer tumor suppressor, but the mechanisms of this suppression are not completely characterized. To determine whether PPARγ regulates MSC expansion in mammary cancer, we deleted PPARγ expression in the mammary epithelium of an in vivo model of basal breast cancer. Loss of PPARγ expression reduced tumor latency, and expanded the CD24+/CD49f(hi) MSC population. PPARγ-null mammary tumors exhibited increased angiogenesis, which was detected in human breast cancer. In vivo inhibition of a PPARγ-regulated miR-15a/angiopoietin-1 pathway blocked increased angiogenesis and MSC expansion. PPARγ bound and activated a canonical response element in the miR-15a gene. PPARγ-null tumors were sensitive to the targeted anti-angiogenic drug sunitinib but resistant to cytotoxic chemotherapy. Normalization of tumor vasculature with sunitinib resulted in objective response to cytotoxic chemotherapy. Chemotherapy-treated PPARγ-null mammary tumors exhibited luminal phenotype and expansion of unipotent CD61+ luminal progenitor cells. Transplantation of chemotherapy-treated luminal progenitor cells recapitulated the luminal phenotype. These results have important implications for anti-angiogenic therapy in breast cancer patients.


Asunto(s)
Células Madre Neoplásicas/patología , PPAR gamma/metabolismo , Nicho de Células Madre , Animales , Eliminación de Gen , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , PPAR gamma/deficiencia , PPAR gamma/genética
2.
Oncogene ; 34(5): 611-20, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-24531712

RESUMEN

Chromosome ends are protected by telomeres that prevent DNA damage response and degradation. Telomerase expression extends telomeres and inhibits DNA damage response. Telomeres are also maintained by the recombination-based alternative lengthening pathway. Telomerase is believed to be the sole mechanism for telomere maintenance in the epidermis. We show that basal cells in the epidermis maintain telomeres both by telomerase and alternative lengthening of telomere (ALT) mechanisms in vivo. ALT was detected in epidermal stem cells in Terc(-/-) mice, and normal human epidermal keratinocytes are also ALT-positive. The ALT pathway is suppressed in primary, but not metastatic, epidermal squamous cell carcinomas (SCC) in Terc(+/+) mice. The ALT pathway is expressed in stem cells and basal cells in epidermal SCC in Terc(-/-) mice, and in some telomerase-positive human SCC lines. Telomeres shorten markedly in stem cells and basal cells in epidermal SCC in vivo. Telomere shortening is associated with telomeric DNA damage response and apoptosis in stem cells and basal cells. Stem cells were transformed in both primary and metastatic epidermal SCC. Genetic ablation of this small cell population resulted in significant tumor regression in vivo. We concluded that alternative lengthening of telomeres is important in epidermal homeostasis and tumorigenesis in vivo.


Asunto(s)
Neoplasias/genética , Células Madre Neoplásicas/patología , ARN/genética , Telomerasa/genética , Homeostasis del Telómero/genética , Telómero/genética , Animales , Apoptosis/genética , Línea Celular , Cromosomas/genética , Daño del ADN/genética , Humanos , Ratones , Neoplasias/patología , Recombinación Genética , Acortamiento del Telómero/genética
3.
Oncogene ; 32(43): 5156-66, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-23178498

RESUMEN

To examine the role of telomeric repeat-binding factor 2 (TRF2) in epithelial tumorigenesis, we characterized conditional loss of TRF2 expression in the basal layer of mouse epidermis. These mice exhibit some characteristics of dyskeratosis congenita, a human stem cell depletion syndrome caused by telomere dysfunction. The epidermis in conditional TRF2 null mice exhibited DNA damage response and apoptosis, which correlated with stem cell depletion. The stem cell population in conditional TRF2 null epidermis exhibited shorter telomeres than those in control mice. Squamous cell carcinomas induced in conditional TRF2 null mice developed with increased latency and slower growth due to reduced numbers of proliferating cells as the result of increased apoptosis. TRF2 null epidermal stem cells were found in both primary and metastatic tumors. Despite the low-grade phenotype of the conditional TRF2 null primary tumors, the number of metastatic lesions was similar to control cancers. Basal cells from TRF2 null tumors demonstrated extreme telomere shortening and dramatically increased numbers of telomeric signals by fluorescence in situ hybridization due to increased genomic instability and aneuploidy in these cancers. DNA damage response signals were detected at telomeres in TRF2 null tumor cells from these mice. The increased genomic instability in these tumors correlated with eightfold expansion of the transformed stem cell population compared with that in control cancers. We concluded that genomic instability resulting from loss of TRF2 expression provides biological advantages to the cancer stem cell population.


Asunto(s)
Daño del ADN/genética , Acortamiento del Telómero , Telómero/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Animales , Carcinogénesis , Inestabilidad Genómica , Humanos , Hibridación Fluorescente in Situ , Ratones , Ratones Noqueados , Células Madre Neoplásicas , Proteína 2 de Unión a Repeticiones Teloméricas/biosíntesis
4.
Histol Histopathol ; 19(2): 505-9, 2004 04.
Artículo en Inglés | MEDLINE | ID: mdl-15024711

RESUMEN

Stem cells have been shown to exist in a variety of tissues. Recent studies have characterized stem cell gene expression patterns, phenotypes, and potential therapeutic uses. One of the most important properties of stem cells is that of self renewal. This raises the possibility that some of the clinical properties of human tumors may be due to transformed stem cells. Similar signaling pathways may regulate self renewal in normal and transformed stem cells. These rare transformed stem cells may drive the process of tumorigenesis due to their potential for self renewal. There are important ramifications for clinical cancer treatment if the growth of solid tumors is at least partially dependent on a cancer stem cell population. In the cancer stem cell model, tumor recurrence may be due to the non-targeted stem cell compartment repopulating the tumor. If cancer stem cells can be prospectively identified and isolated, it should be possible to identify therapies that will selectively target these cells.


Asunto(s)
Neoplasias/metabolismo , Células Madre/citología , Animales , Línea Celular Transformada , Línea Celular Tumoral , Linaje de la Célula , Transformación Celular Neoplásica , Humanos , Ratones , Fenotipo , Transducción de Señal
5.
Am J Physiol Lung Cell Mol Physiol ; 283(4): L700-6, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12225946

RESUMEN

Murine Sprouty2 (mSpry2) is a conserved ortholog of Drosophila Sprouty, a gene that inhibits several tyrosine kinase receptor pathways, resulting in net reduction of mitogen-activated protein (MAP) kinase activation. However, the precise mechanism mediating mSpry2 function as a negative regulator in tyrosine kinase growth factor pathways that regulate diverse biological functions remains incompletely characterized. Fibroblast growth factor 10 (FGF10) is a key positive regulator of lung branching morphogenesis and induces epithelial expression of mSpry2 adjacent to mesenchymal sites of FGF10. Herein, we demonstrate that FGF10 stimulation of mouse lung epithelial cells (MLE15) overexpressing mSpry2 results in both mSpry2 tyrosine phosphorylation and differential binding of mSpry2 to several key upstream target proteins in the MAP kinase-activating pathway. Thus FGF receptor (FGFR) activation results in increased association of mSpry2 with growth factor receptor-binding protein 2, suc-1-associated nuerotrophic factor target 2, and Raf but decreased binding to protein tyrosine phosphatase 2 and GTPase-activating protein 1, resulting in a net reduction of MAP kinase activation. mSpry2 also spatially translocates to the plasma membrane and intracellular membrane structures in response to FGF10 stimulation. Our data demonstrate novel intracellular mechanisms mediating mSpry2 function as a negative regulator of uncontrolled FGF-induced MAP kinase signaling.


Asunto(s)
Factores de Crecimiento de Fibroblastos/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/farmacología , Mucosa Respiratoria/metabolismo , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Células Cultivadas , Expresión Génica/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Fosforilación , Mucosa Respiratoria/citología , Tirosina/metabolismo
6.
Histol Histopathol ; 17(3): 909-14, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12168802

RESUMEN

Squamous cell carcinoma of the head and neck region (HNSCC) is the sixth most frequent cancer worldwide, comprising almost 50% of all malignancies in some developing nations. In the United States, 30,000 new cases and 8,000 deaths are reported each year. Survival rates vary depending on tobacco and alcohol consumption, age, gender, ethnic background, and geographic area. This variability reflects the multifactorial pathogenesis of the disease. Early detection and diagnosis has increased survival but the overall 5 year rate of 50% is among the lowest of the major cancers. Differences between normal epithelium and cancer cells of the upper aerodigestive tract arise from specific alterations in genes controlling DNA repair, proliferation, immortalization, apoptosis, invasion, and angiogenesis. These proteins include both tumor suppressors and activating oncogenes which regulate a wide variety of intracellular signaling pathways. Included in these pathways are growth factor receptors, signal transducers, and transcription factors which regulate DNA damage response, cell cycle arrest, and programmed cell death. In head and neck cancer, alterations of three signaling pathways occur with sufficient frequency and produce such dramatic phenotypic changes as to be considered the critical transforming events of the disease. These changes include mutation of the p53 tumor suppressor, inactivation of the cyclin dependent kinase inhibitor p16, and overexpression of epidermal growth factor receptor (EGFR). This review will focus on the molecular changes which occur in these pathways and how they contribute to the pathogenesis of HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Receptores ErbB/genética , Genes Supresores de Tumor , Genes p53/genética , Humanos , Modelos Biológicos , Mutación
7.
Curr Cancer Drug Targets ; 2(1): 77-86, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12188922

RESUMEN

For many years, the vitamin A metabolite retinoic acid (RA) has been known to have profound effects on development, cellular proliferation and differentiation, and tumor growth and invasion. The wide-ranging effects of RA on cellular proliferation and migration have made it a useful chemotherapeutic agent in the treatment of many types of cancer. In the last fifteen years, with the discovery of nuclear receptors for RA, the molecular basis for the effects of this molecule has become apparent. Retinoic acid receptors (RAR) are members of a superfamily of ligand dependent transcription factors that interact with an increasingly large array of coactivators and repressors to regulate target gene expression through binding to cognate recognition sequences in the promoters of these genes. Alterations in RAR expression and function have been demonstrated in many types of cancer. The translocation of RARalpha with PML or PLZF genes in acute promyelocytic leukemia is a paradigm of the role of RARs in cancer biology. In addition, the development of receptor selective synthetic retinoids has greatly expanded our knowledge of RAR function in tumor cells and provided additional treatment options for cancer patients. This review will examine the development of receptor selective retinoids, their uses to date, and future potential.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Receptores de Ácido Retinoico/metabolismo , Retinoides/síntesis química , Retinoides/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Humanos , Neoplasias/metabolismo , Retinoides/metabolismo
8.
Mol Carcinog ; 32(4): 187-94, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11746830

RESUMEN

Growth factor receptors of the tyrosine kinase family regulate proliferation of a variety of cell types. In some human cancers, the epidermal growth factor receptor (EGFR) and its ligands often are overexpressed, leading to both constitutive and autocrine activation. Intracellular signaling via this receptor takes place through several mechanisms of action, including activation of ras and the mitogen-activated protein kinase (MAPK) pathway. Our previous studies have shown that human squamous cell carcinoma (SCC) lines overexpress EGFR and do not increase proliferation in response to exogenous epidermal growth factor (EGF). The vitamin A metabolite retinoic acid (RA) has been used as a chemotherapeutic drug in the treatment of SCC. RA decreases proliferation of SCC lines, in part owing to inhibition of EGFR expression. However, we previously found that treatment of SCC lines with inhibitory doses of RA sensitized cells to the proliferative effects of EGF. We now present a mechanism of action for this effect. RA inhibited expression of EGFR and proteins in the MAPK signaling pathway. Expression of these molecules returned to basal levels within 24 h after RA withdrawal. RA also inhibited autocrine secretion of EGF, which returned to basal levels with slower kinetics. During this time, addition of exogenous EGF stimulated mitosis in SCC lines. These data suggested that signaling proteins downstream of overexpressed EGFR may have limited the mitotic response in SCC lines. In support of this hypothesis, overexpression of the EGFR adaptor protein Grb2 increased cell proliferation and restored EGF-induced mitosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/biosíntesis , Proteínas/metabolismo , Tretinoina/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , División Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Proteína Adaptadora GRB2 , Humanos , Transducción de Señal/efectos de los fármacos , Transfección , Tretinoina/uso terapéutico , Células Tumorales Cultivadas
9.
Nucleic Acids Res ; 29(13): 2789-94, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11433024

RESUMEN

The ends of human chromosomes (telomeres) lose up to 200 bp of DNA per cell division. Chromosomal shortening ultimately leads to senescence and death in normal cells. Many human carcinoma lines are immortal in vitro, suggesting that these cells have a mechanism for maintaining the ends of their chromosomes. Telomerase is a ribonucleoprotein complex that synthesizes telomeric DNA onto chromosomes using its RNA component as template. Telomerase activity is found in most tumor cells, but is absent from normal cells. Little is known about how normal human cells repress telomerase (hTERT) gene expression. Mice carrying an E2F-1 null mutation develop a variety of malignant tumors, suggesting that this transcription factor has a tumor suppressor function. To determine mechanisms by which E2F-1 suppresses tumor formation, we examined the role of this transcription factor in regulation of the hTERT promoter in human cells. We identified two putative E2F-1-binding sites proximal to the transcriptional start site of the hTERT promoter. Mutation of these sites produced dramatic increases in promoter activity. Overexpression of E2F-1 but not a mutant E2F-1 repressed hTERT promoter activity in reporter gene assays. This repression was abolished by mutation of the E2F-1-binding sites in the hTERT promoter. Human cancer cell lines stably overexpressing E2F-1 exhibited decreased hTERT mRNA expression and telomerase activity. We conclude that E2F-1 has an atypical function as a transcriptional repressor of the hTERT gene in human cells.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Unión al ADN/metabolismo , Regulación Enzimológica de la Expresión Génica , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Telomerasa/genética , Factores de Transcripción/metabolismo , Secuencia de Bases , Sitios de Unión , Secuencia de Consenso/genética , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/genética , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Regulación Neoplásica de la Expresión Génica , Genes Reporteros/genética , Genes Supresores de Tumor/genética , Humanos , Datos de Secuencia Molecular , Mutación/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteína 1 de Unión a Retinoblastoma , Telomerasa/metabolismo , Factores de Transcripción/genética , Células Tumorales Cultivadas
10.
Neoplasia ; 3(1): 27-32, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11326313

RESUMEN

Tumor cell invasion and metastasis require precise coordination of adherence to extracellular matrix (ECM) and controlled degradation of its components. Invasive cells secrete proteolytic enzymes known as matrix metalloproteinases (MMPs) which degrade specific basement membrane molecules. Expression of these enzymes is regulated by multiple signaling mechanisms, including the mitogen-activated protein kinase (MAPK) pathway. One of the terminal effectors of this signaling cascade is jun N-terminal kinase 1 (JNK1) which phosphorylates the transcription factor c-jun, a component of the AP-1 complex. MMP-9 expression is regulated by two well-characterized AP-1 sites in the promoter of this gene. To determine how JNK1 activity regulated MMP-9 expression in human squamous cell carcinoma lines, we overexpressed this kinase in SCC25 cells. JNK1 overexpression induced MMP-9 protein levels and activity in this cell line. Elevated MMP-9 expression correlated with increased invasion of reconstituted basement membranes by JNK1-overexpressing clones. Site-directed mutagenesis of the MMP-9 promoter revealed that JNK1 cooperated with its transcription factor target c-jun to increase MMP-9 expression at the transcriptional level via the proximal AP-1 site. These results suggest that elevated JNK1 expression may contribute to increased MMP-9 activity and ECM invasion by tumor cells.


Asunto(s)
Metaloproteinasa 9 de la Matriz/genética , Proteínas Quinasas Activadas por Mitógenos/fisiología , Regiones Promotoras Genéticas , Transcripción Genética , Sitios de Unión , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Cloranfenicol O-Acetiltransferasa/metabolismo , Matriz Extracelular/metabolismo , Humanos , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína Quinasa 8 Activada por Mitógenos , Invasividad Neoplásica , Pruebas de Precipitina , ARN Mensajero/biosíntesis , Factor de Transcripción AP-1/metabolismo , Transfección , Células Tumorales Cultivadas , Regulación hacia Arriba
11.
Biochim Biophys Acta ; 1518(1-2): 1-6, 2001 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-11267653

RESUMEN

The ends of human chromosomes (telomeres) lose up to 200 bp of DNA per cell division. Chromosomal shortening ultimately leads to senescence and death in normal cells. Many human carcinoma lines are immortal in vitro, suggesting that these cells have a mechanism for maintaining the ends of their chromosomes. Telomerase is a ribonucleoprotein complex that synthesizes telomeric DNA onto chromosomes using its RNA component as a template. Recent studies have shown that inactivation of the retinoblastoma gene product pRb and the cyclin dependent kinase inhibitor p16(INK4A) is required for telomerase activity in epithelial cells. We have demonstrated previously that restoration of functional retinoblastoma (Rb) expression is sufficient to downregulate telomerase activity in carcinoma cells. To determine mechanisms by which Rb regulates telomerase expression, we examined the effects of cyclin dependent kinase (cdk) mediated Rb inactivation and the release of E2F-1 on telomerase activity in human carcinoma cells. Overexpression of cdk2 and cdk4 but not a dominant negative cdk2 rescued Rb mediated downregulation of telomerase activity. Overexpression of the cdk regulatory subunit cyclin D1 also rescued telomerase downregulation and p16 expression alone was sufficient to ablate activity. E2F-1 overexpression was sufficient to rescue Rb mediated reduction of telomerase activity, but an E2F-1 mutant defective in DNA and Rb binding activities failed to produce this effect. Tumor tissue from E2F-1 -/- mice was negative for telomerase activity, indicating a key regulatory role for this transcription factor.


Asunto(s)
Quinasas CDC2-CDC28 , Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Proteínas Proto-Oncogénicas , Proteína de Retinoblastoma/metabolismo , Telomerasa/metabolismo , Factores de Transcripción/metabolismo , Animales , Ciclina D1/biosíntesis , Ciclina D1/genética , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/biosíntesis , Quinasas Ciclina-Dependientes/genética , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Humanos , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteína de Retinoblastoma/biosíntesis , Proteína de Retinoblastoma/genética , Proteína 1 de Unión a Retinoblastoma , Factor de Transcripción DP1 , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Células Tumorales Cultivadas
12.
Int J Oncol ; 18(2): 369-74, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11172606

RESUMEN

Tumor cell invasion and metastasis require precise coordination of adherence to extracellular matrix (ECM) and controlled degradation of its components. Invasive cells secrete proteolytic enzymes known as matrix metalloproteinases (MMPs) which degrade specific basement membrane molecules. Expression of these enzymes is regulated by multiple signaling mechanisms, including ECM attachment itself. Previously, we have shown that retinoic acid (RA) inhibits tumor cell invasion of ECM by regulating MMP expression. We determined that RA mediated inhibition of MMP-9 expression was dependent on ECM attachment in RA sensitive but not RA resistant human carcinoma lines. In the present study we examined the mechanisms of this regulation. Both RA and attachment to type IV collagen inhibited the activity of one of the terminal effectors of the mitogen activated protein kinase (MAPK) pathway, namely ERK1. RA also induced ets-1 expression, a transcription factor known to be phosphorylated by ERK1, in RA sensitive but not RA resistant lines. Increased expression of hypophosphorylated ets-1 was sufficient to inhibit MMP-9 expression in these lines. This inhibition was mediated via two ets-1 binding sites in the MMP-9 promoter. The results of these experiments indicate that induction of hypophosphorylated ets-1 as the result of RA and ECM mediated decreases in ERK1 activity represents a novel mechanism by which RA regulates MMP-9 gene expression.


Asunto(s)
Antineoplásicos/farmacología , Matriz Extracelular/enzimología , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Proto-Oncogénicas/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos , Tretinoina/farmacología , Genes Reporteros/fisiología , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Proto-Oncogénica c-ets-1 , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
13.
Anticancer Res ; 20(5A): 2945-9, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11062705

RESUMEN

The mitogen activated protein kinase (MAPK) pathway is a paradigm for regulation of growth factor signaling and cellular proliferation. The MAPK pathway is a major target for signaling by growth factor receptor kinases. The MAPK pathway consists of a series of protein kinases which is activated by phosphorylation of specific amino acid residues in their regulatory domains. The MAPK family can be divided into three subgroups: the extracellular signal regulated kinases (ERKs), the stress activated protein kinase/jun N terminal kinase (SAPK/JNK), and the p38 MAPK. These kinase cascades phosphorylate transcription factor targets such as ets, c-jun, and ATF-2. Of these, little is known about the role of ATF-2 in regulation of MAPK signaling and cellular proliferation. To begin to understand this role, we overexpressed ATF-2 in a human cancer cell line. ATF-2 inhibited the G1/S phase transition of the cell cycle and decreased the proliferation rate of these cells. Decreased proliferation correlated with cell cycle independent inhibition of ERK1 expression in ATF-2 clones. Genetic and pharmacologic inhibition of ERK1 activity was sufficient to reproduce the effects of ATF-2 on cell cycle progression and proliferation. These results indicate a novel role for ATF-2 in cancer cell proliferation and suggest a potential feedback mechanism that regulates MAPK signaling.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factores de Transcripción/metabolismo , Factor de Transcripción Activador 2 , Proteína Quinasa CDC2/metabolismo , Carcinoma de Células Escamosas , Ciclo Celular , División Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Humanos , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Factores de Transcripción/genética , Células Tumorales Cultivadas
14.
Oral Oncol ; 36(1): 61-6, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10889921

RESUMEN

Angiogenesis is the formation of capillaries from pre-existing blood vessels. Angiogenesis occurs in many developmental, physiologic, and pathologic processes including tumor growth. Previous studies have shown that angiogenesis is required for growth and metastasis of solid tumors. Fibroblast growth factors (FGF-1, -2) and vascular endothelial growth factor (VEGF) are extremely potent angiogenesis inducers by stimulating the proliferation and migration of capillary endothelial cells. Expression of these factors is upregulated in many solid tumors and correlates with high vascularity, lymph node metastasis, and poor clinical prognosis. Few studies have examined whether established head and neck squamous cell carcinoma (SCC) lines produce biologically active angiogenic factors. By immunoprecipitation, we detected FGF and VEGF proteins in cellular lysates of SCC lines. We also detected FGF-1 and -2 proteins in serum-free-conditioned medium from these lines. Conditioned medium from SCC lines significantly increased proliferation of human umbilical vein endothelial cells (HUVEC). This increased proliferation was abrogated by pre-incubation of conditioned medium with neutralizing antibodies to FGFs and VEGF. Conditioned medium from SCC lines also significantly stimulated HUVEC invasion across a reconstituted basement membrane. We concluded that head and neck SCC lines secrete biologically active angiogenic factors.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Proteínas de Neoplasias/metabolismo , Factores de Crecimiento Endotelial/metabolismo , Endotelio Vascular/patología , Ensayo de Inmunoadsorción Enzimática , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Linfocinas/metabolismo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
15.
Mol Cell Biol Res Commun ; 3(4): 243-8, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10891399

RESUMEN

c-fos is the prototypic member of a family of transcription factors that regulate many cellular processes, including proliferation. c-fos heterodimerizes with jun family members to form the AP-1 transcription factor complex which binds specific DNA recognition elements in the promoters of many genes. Following rapid induction in response to serum or growth factors, c-fos regulates expression of downstream target genes involved in cellular proliferation. Although much work has focused on activation of cell cycle regulatory genes by c-fos, less is known about negative regulation of gene expression by this transcription factor. The cyclin-dependent kinase (cdk) inhibitor p21(Cip1/WAF1) is a negative regulator of cdk activity, thereby impeding cell cycle progression. By sequence analysis, we identified a putative AP-1 element in the p21(Cip1/WAF1) promoter. To investigate how this site regulated p21(Cip1/WAF1) expression and mitigate external effects on c-fos expression, we used a c-fos/estrogen receptor (c-fosER) fusion construct in which this transcription factor is conditionally activated by estradiol. In the presence of estradiol, c-fosER downregulated p21(Cip1/WAF1) promoter activity. This inhibition was dependent on the putative AP-1 site. Activation of c-fosER induced cell cycle progression and proliferation in a manner similar to serum stimulation. We concluded that activation of c-fosER mediated transcriptional inhibition of p21(Cip1/WAF1) through a previously uncharacterized AP-1 site, revealing an important role for c-fos in negative control of cell cycle regulatory genes.


Asunto(s)
Ciclo Celular , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Estradiol/farmacología , Genes Reporteros , Humanos , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-fos/genética , Receptores de Estrógenos/genética , Proteínas Recombinantes de Fusión/genética , Elementos de Respuesta/genética , Factores de Tiempo , Factor de Transcripción AP-1/fisiología , Activación Transcripcional/efectos de los fármacos , Transfección , Células Tumorales Cultivadas
16.
Mech Dev ; 93(1-2): 71-81, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10781941

RESUMEN

Smad7 was recently shown to antagonize TGF-beta-induced activation of signal-transducing Smad2 and Smad3 proteins. However, the biological function of Smad7 in the process of lung organogenesis is not known. Since Smad2/3-mediated TGF-beta signaling is known to inhibit embryonic lung branching morphogenesis, we tested the hypothesis that Smad7 regulates early lung development by modulating TGF-beta signal transduction. An antisense oligodeoxynucleotide (ODN) was designed to specifically block endogenous Smad7 gene expression at both transcriptional and translational levels in embryonic mouse lungs in culture. TGF-beta-mediated inhibition of lung branching morphogenesis was significantly potentiated in cultured embryonic lungs in the absence of Smad7 gene expression: abrogation of Smad7 potentiated TGF-beta-mediated inhibition of lung branching morphogenesis from 76 to 52% of the basal level in lungs cultured in the presence of 5 ng/ml TGF-beta1 ligand. Likewise, TGF-beta1 EC(50) (concentration of TGF-beta1 that induced half maximal branching inhibition) was reduced from 5 to 1 ng/ml when Smad7 gene expression was abrogated in lung culture, indicating an enhanced level of TGF-beta signaling in lung tissue with abolished Smad7 gene expression. By immunocytochemistry, Smad7 protein was co-localized with both Smad2 and Smad3 in distal bronchial epithelial cells, supporting the concept that Smad7 inhibits TGF-beta signaling by competing locally with Smad2 and Smad3 for TGF-beta receptor complex binding during lung morphogenesis. Furthermore, antisense Smad7 ODN increased the negative effect of TGF-beta1 on epithelial cell growth in developing lungs in culture. We also demonstrated that Smad7 mRNA levels were rapidly and potently induced upon TGF-beta1 stimulation of lungs in culture, suggesting that Smad7 regulates TGF-beta responses in a negative feedback loop. These studies define a novel function for Smad7 as an intracellular antagonist of TGF-beta-induced, Smad2/3-mediated inhibition of murine embryonic lung growth and branching morphogenesis in culture. The optimization of TGF-beta signaling during early lung development therefore requires a finely-regulated competitive balance between both permissive and inhibitory members of the Smad family.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/metabolismo , Pulmón/embriología , Transactivadores/biosíntesis , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , División Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Epitelio/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Pulmón/metabolismo , Ratones , Oligodesoxirribonucleótidos Antisentido , Técnicas de Cultivo de Órganos , ARN Mensajero , Proteína Smad2 , Proteína smad3 , Proteína smad7 , Transactivadores/genética , Factor de Crecimiento Transformador beta/farmacología
17.
IUBMB Life ; 50(6): 365-70, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11327309

RESUMEN

The vitamin A metabolite retinoic acid (RA) is a powerful regulator of cellular proliferation and differentiation. The effects of RA on target gene expression are mediated by a family of ligand dependent nuclear transcription factors known as retinoic acid receptors (RAR). RARs have functional domains for retinoic acid binding, dimerization, and transactivation. RA response elements (RARE) found in the promoters of many genes consist of variable direct repeats of the sequence PuGGTCA spaced by five nucleotides (DR5). We have identified a novel DR5 element in the human ets-1 promoter. Mutational analysis of this site indicates that it is necessary for RA- and RAR-dependent activation of the ets-1 promoter. RARalpha can bind specifically to this site as determined by electrophoretic mobility shift analysis. Finally, RA mediates induction of the human ets-1 gene at the mRNA and protein levels. These data suggest that induction of ets-1 expression by RA is mediated by a novel retinoic acid response element in the promoter region of this gene.


Asunto(s)
Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Elementos de Respuesta/genética , Factores de Transcripción/genética , Tretinoina/metabolismo , Células Cultivadas , Análisis Mutacional de ADN , Electroforesis , Humanos , Proteína Proto-Oncogénica c-ets-1 , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-ets , Receptores de Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Análisis de Secuencia de ADN , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Tretinoina/farmacología
18.
Int J Oncol ; 15(3): 519-23, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10427134

RESUMEN

Growth factor receptors of the tyrosine kinase family regulate proliferation and migration of a variety of cell types. Binding of cognate ligands to these receptors induces multiple cellular responses, including cell cycle progression and motility in culture model systems. In stratified squamous epithelial cells, these receptors include epidermal growth factor receptor (EGFR) that binds both EGF and transforming growth factor alpha (TGFalpha), and c-met whose ligand is hepatocyte growth factor/scatter factor (HGF/SF). Intracellular signaling via these receptors occurs by several mechanisms, including activation of ras, phosphatidylinositol 3-kinase (PI3K), and the mitogen activated protein kinase (MAPK) pathways. Growth factor independence is a characteristic feature of transformation in cancer cells. Previous studies have shown that human squamous cell carcinoma (SCC) lines do not require EGF or TGFalpha for proliferation. We show that while these cell lines expressed EGFR and c-met, stimulation with their respective ligands did not induce proliferation but markedly increased invasion of reconstituted basement membranes. However, EGFR kinase activity was required for proliferation and EGF induced invasion by these cells. Signaling via ras, PI3K, and MAPK was required for proliferation of SCC lines. However, inhibition of ras and MAPK did not significantly reduce invasion by these cells nor completely block stimulation of this activity by EGF and HGF. We concluded that MAPK signaling was required for proliferation but not invasion of human SCC lines.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Carcinoma de Células Escamosas/enzimología , Receptores de Factores de Crecimiento/fisiología , Carcinoma de Células Escamosas/patología , División Celular/fisiología , Humanos , Invasividad Neoplásica , Transducción de Señal/fisiología , Células Tumorales Cultivadas
19.
J Dent Res ; 78(6): 1256-63, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10371250

RESUMEN

Squamous cell carcinoma (SCC) of the head and neck is the sixth most frequent cancer worldwide. The survival rate is among the lowest of the major cancers and has not improved significantly in the past two decades. Extensive local invasion and regional lymph node metastasis are, in large part, responsible for the poor clinical outcome of these tumors. Keratin intermediate filaments are the most abundant cytoskeletal proteins in SCCs and regulate the migration of normal and transformed epithelial cells. Previous studies have shown that expression of the 40-kDa keratin K19 is dysregulated in SCCs arising from oral epithelium. Immunohistochemical experiments demonstrated that, while normal epithelium and dysplastic lesions expressed abundant K19 protein, invasive SCCs exhibited a patchy or negative staining pattern. We subsequently determined that K19 expression was consistently downregulated in seven SCC lines compared with normal epithelium. We therefore wanted to determine if K19 downregulation affected the invasive phenotype of these cells. We found that SCC lines which do not express K19 are significantly more invasive in vitro than those which retain expression of this gene. Stable expression of the K19 cDNA in K19 negative cell lines altered cell morphology and intercellular adhesiveness, and significantly decreased the number of cells able to migrate through a reconstituted basement membrane. Reduced invasiveness was not due to decreased metalloproteinase activity in the K19-expressing clones. We conclude that K19 overexpression in oral SCCs decreases their invasive potential by diminishing migratory capability.


Asunto(s)
Carcinoma de Células Escamosas/patología , Regulación hacia Abajo , Queratinas/genética , Neoplasias de la Boca/patología , Membrana Basal/patología , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/secundario , Adhesión Celular/genética , Línea Celular Transformada , Movimiento Celular/genética , Colorantes , ADN Complementario/genética , Electroforesis en Gel de Poliacrilamida , Células Epiteliales/patología , Técnica del Anticuerpo Fluorescente , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Proteínas de Filamentos Intermediarios/genética , Metástasis Linfática/patología , Metaloendopeptidasas/genética , Mucosa Bucal/patología , Neoplasias de la Boca/genética , Invasividad Neoplásica , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas
20.
Biochim Biophys Acta ; 1445(2): 207-15, 1999 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-10320773

RESUMEN

The ends of human chromosomes (telomeres) consist of tandem repeats of the sequence TTAGGG. Telomeres lose up to 200 base pairs of DNA per cell division due to the inability of DNA polymerase to completely replicate the chromosomal ends. Chromosomal shortening ultimately leads to senescence and cell death in normal cells. However, some immortal cells do not lose telomeric sequence during DNA replication. Many human carcinoma lines are immortal in vitro, suggesting that these cells have a mechanism for maintaining the ends of their chromosomes. Telomerase is a ribonucleoprotein complex that synthesizes telomeric DNA onto chromosomes using its RNA component as a template. To elucidate potential mechanisms for telomerase regulation, we tested human squamous cell carcinoma lines (SCCs) for telomerase activity. All SCC lines expressed high levels of telomerase activity. Synchronization in specific cell cycle phases caused marked reduction in telomerase activity in G0 and S, but not in G1 or M. Reduction in telomerase activity correlated with induction of Rb protein in these phases. Overexpression of full length Rb resulted in significant downregulation of telomerase activity. However, expression of an Rb N-terminal oligomerization domain deletion construct, a C-terminal DNA binding domain deletion construct, or a pocket domain mutant failed to downregulate telomerase activity. We concluded that functionally intact Rb was required for cell cycle-dependent downregulation of telomerase activity in SCC lines.


Asunto(s)
Proteína de Retinoblastoma/genética , Telomerasa/genética , Regulación hacia Abajo , Citometría de Flujo , Expresión Génica , Humanos , Mutación , Proteína de Retinoblastoma/biosíntesis , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA