Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Acta Biomed ; 90(11-S): 29-37, 2019 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-31714498

RESUMEN

BACKGROUND AND AIM OF THE STUDY: Working in the emergency medical service often exposes nurses to highly stressful situations and can impact their quality of life. Among the strategies aimed at mitigating the effects of this phenomenon, peer-supporting represents an emerging model used in the emergency medical service setting. The aim of the study is to explore the experiences, the opinions and feelings of emergency medical service nursing staff in relation to the use of the peer supporting model. METHODS: A semi-structured interview was carried out. Participants were recruited on a voluntary basis from an emergency medical service in the north of Italy. Interviews were audio-recorded and the data extracted were anonymised. RESULTS: 14 nurses participated in the study. The totality of the participants recognized that their daily clinical practice, especially when involving paediatric patients, can have a profound emotional impact on their life in general. Furthermore, interviewees admitted that their personal copying mechanisms did not seem to be entirely effective when processing their painful experiences.  The majority of the participants were in favour of introducing a peer-supporter in the ambulance service. CONCLUSIONS: This study emphasises the need to implement emotional support tools for non-hospital emergency nurses in daily clinical practice, in order to facilitate emotional decompression secondary to particularly stressful interventions as soon as possible. The peer-supporting strategy could represent, in this direction, a valid and shared model.


Asunto(s)
Adaptación Psicológica , Ambulancias , Personal de Enfermería en Hospital/psicología , Femenino , Humanos , Masculino , Grupo Paritario , Autocuidado
2.
Carcinogenesis ; 36(10): 1180-92, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26168819

RESUMEN

There is a well-established association between aging and the onset of metastasis. Although the mechanisms through which age impinges upon the malignant phenotype remain uncharacterized, the role of a senescent microenvironment has been emphasized. We reported previously that human epithelial cells that undergo telomere-driven chromosome instability (T-CIN) display global microRNA (miR) deregulation and develop migration and invasion capacities. Here, we show that post-crisis cells are not able to form tumors unless a senescent microenvironment is provided. The characterization of cell lines established from such tumors revealed that these cells have acquired cell autonomous tumorigenicity, giving rise to heterogeneous tumors. Further experiments demonstrate that explanted cells, while displaying differences in cell differentiation markers, are all endowed of enhanced stem cell properties including self-renewal and multilineage differentiation capacity. Treatments of T-CIN+ cells with senescence-conditioned media induce sphere formation exclusively in cells with senescence-associated tumorigenicity, a capacity that depends on miR-145 repression. These results indicate that the senescent microenvironment, while promoting further transdifferentiations in cells with genome instability, is able to propel the progression of premalignant cells towards a malignant, cell stem-like state.


Asunto(s)
Envejecimiento/genética , Transformación Celular Neoplásica/genética , MicroARNs/biosíntesis , Células Madre Neoplásicas/patología , Microambiente Tumoral/genética , Envejecimiento/patología , Diferenciación Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Senescencia Celular/genética , Inestabilidad Cromosómica/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Telómero/genética
3.
PLoS One ; 8(5): e62522, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23658738

RESUMEN

In previous work we have documented the nuclear translocation of endothelial NOS (eNOS) and its participation in combinatorial complexes with Estrogen Receptor Beta (ERß) and Hypoxia Inducible Factors (HIFs) that determine localized chromatin remodeling in response to estrogen (E2) and hypoxia stimuli, resulting in transcriptional regulation of genes associated with adverse prognosis in prostate cancer (PCa). To explore the role of nuclear eNOS in the acquisition of aggressive phenotype in PCa, we performed ChIP-Sequencing on chromatin-associated eNOS from cells from a primary tumor with poor outcome and from metastatic LNCaP cells. We found that: 1. the eNOS-bound regions (peaks) are widely distributed across the genome encompassing multiple transcription factors binding sites, including Estrogen Response Elements. 2. E2 increased the number of peaks, indicating hormone-dependent eNOS re-localization. 3. Peak distribution was similar with/without E2 with ≈ 55% of them in extragenic DNA regions and an intriguing involvement of the 5' domain of several miRs deregulated in PCa. Numerous potentially novel eNOS-targeted genes have been identified suggesting that eNOS participates in the regulation of large gene sets. The parallel finding of downregulation of a cluster of miRs, including miR-34a, in PCa cells associated with poor outcome led us to unveil a molecular link between eNOS and SIRT1, an epigenetic regulator of aging and tumorigenicity, negatively regulated by miR-34a and in turn activating eNOS. E2 potentiates miR-34a downregulation thus enhancing SIRT1 expression, depicting a novel eNOS/SIRT1 interplay fine-tuned by E2-activated ER signaling, and suggesting that eNOS may play an important role in aggressive PCa.


Asunto(s)
Carcinoma/genética , Estradiol/metabolismo , Receptor beta de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias de la Próstata/genética , Sirtuina 1/genética , Carcinoma/diagnóstico , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Estradiol/farmacología , Receptor beta de Estrógeno/metabolismo , Retroalimentación Fisiológica , Perfilación de la Expresión Génica , Humanos , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Cultivo Primario de Células , Pronóstico , Regiones Promotoras Genéticas , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Precursores del ARN/genética , Precursores del ARN/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo
4.
Nucleic Acids Res ; 41(9): 4792-801, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23519615

RESUMEN

In contrast with the limited sequence divergence accumulated after separation of higher primate lineages, marked cytogenetic variation has been associated with the genome evolution in these species. Studying the impact of such structural variations on defined molecular processes can provide valuable insights on how genome structural organization contributes to organismal evolution. Here, we show that telomeres on chromosome arms carrying subtelomeric heterochromatic caps in the chimpanzee, which are completely absent in humans, replicate later than telomeres on chromosome arms without caps. In gorilla, on the other hand, a proportion of the subtelomeric heterochromatic caps present in most chromosome arms are associated with large blocks of telomere-like sequences that follow a replication program different from that of bona fide telomeres. Strikingly, telomere-containing RNA accumulates extrachromosomally in gorilla mitotic cells, suggesting that at least some aspects of telomere-containing RNA biogenesis have diverged in gorilla, perhaps in concert with the evolution of heterochromatic caps in this species.


Asunto(s)
Gorilla gorilla/genética , Heterocromatina/química , Pan troglodytes/genética , Telómero/metabolismo , Animales , Línea Celular , Hominidae , Mitosis/genética , ARN/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos , Telómero/química , Transcripción Genética
5.
Carcinogenesis ; 34(5): 1173-80, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23358853

RESUMEN

Telomere shortening is a major source of chromosome instability (CIN) at early stages during carcinogenesis. However, the mechanisms through which telomere-driven CIN (T-CIN) contributes to the acquisition of tumor phenotypes remain uncharacterized. We discovered that human epithelial kidney cells undergoing T-CIN display massive microRNA (miR) expression changes that are not related to local losses or gains. This widespread miR deregulation encompasses a miR-200-dependent epithelial-to-mesenchymal transition (EMT) that confers to immortalized pre-tumoral cells phenotypic traits of metastatic potential. Remarkably, a miR signature of these cells, comprising a downregulation of miRs with conserved expression in kidney, was retrieved in poorly differentiated aggressive renal cell carcinomas. Our results reveal an unanticipated connection between telomere crisis and the activation of the EMT program that occurs at pre-invasive stages of epithelial cancers, through mechanisms that involve miR deregulation. Thus, this study provides a new rational into how telomere instability contributes to the acquisition of the malignant phenotype.


Asunto(s)
Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Neoplasias Renales/genética , Neoplasias Renales/patología , MicroARNs/genética , Telómero/genética , Línea Celular Tumoral , Inestabilidad Cromosómica/genética , Daño del ADN/genética , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Células HEK293 , Humanos , Transcripción Genética/genética
6.
Mol Biol Cell ; 23(8): 1467-74, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22379106

RESUMEN

In vivo imaging involving the use of genetically engineered animals is an innovative powerful tool for the noninvasive assessment of the molecular and cellular events that are often targets of therapy. On the basis of the knowledge that the activity of the nuclear factor-Y (NF-Y) transcription factor is restricted in vitro to proliferating cells, we have generated a transgenic reporter mouse, called MITO-Luc (for mitosis-luciferase), in which an NF-Y-dependent promoter controls luciferase expression. In these mice, bioluminescence imaging of NF-Y activity visualizes areas of physiological cell proliferation and regeneration during response to injury. Using this tool, we highlight for the first time a role of NF-Y activity on hepatocyte proliferation during liver regeneration. MITO-Luc reporter mice should facilitate investigations into the involvement of genes in cell proliferation and provide a useful model for studying aberrant proliferation in disease pathogenesis. They should be also useful in the development of new anti/proproliferative drugs and assessment of their efficacy and side effects on nontarget tissues.


Asunto(s)
Factor de Unión a CCAAT/genética , Factor de Unión a CCAAT/metabolismo , Proliferación Celular , Regeneración Hepática , Imagen Molecular , Transcripción Genética , Animales , Ciclo Celular/genética , Línea Celular , Ciclina B2/genética , Proteínas de Unión al ADN/genética , Genes Reporteros , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas
7.
Horm Mol Biol Clin Investig ; 5(2): 91-6, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25961244

RESUMEN

This review is based on novel observations from our laboratory on the nuclear translocation and functional role of endothelial nitric oxide synthase (eNOS) in endothelial and prostate cancer (PCa) epithelial cells. Nitric oxide (NO), the product of eNOS, is a free radical involved in the physiology and pathophysiology of living organisms and in a variety of biological processes including the maintenance of vascular homeostasis. Of relevance in this context is the role that estrogens play in the apoptotic process and the migration of endothelial cells through the regulation of target genes such as eNOS itself. It has been shown that both estrogen and NO signaling, mediated respectively by the estrogen receptors (ERs) and eNOS, can strongly counteract endothelial senescence through a common effector, the catalytic subunit of human telomerase. Therefore, this protein has been identified as a key molecule in the aging process which, intriguingly, is considered the only risk factor in the development of PCa and one of the major determinants of cardiovascular diseases. Indeed, in both these contexts we have defined a molecular mechanism involving activation of eNOS and hypoxia-inducible factors in association with ERß that characterizes the most aggressive form of PCa or influences endothelial cell differentiation. Altogether these data led us to postulate that activation of eNOS is a crucial requirement for the delaying of endothelial senescence as well as for the acquisition of androgen-independence and for tumor progression in the prostate microenvironment.

8.
Proc Natl Acad Sci U S A ; 106(37): 15726-31, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19717459

RESUMEN

Promyelocytic leukemia (PML) bodies (also called ND10) are dynamic nuclear structures implicated in a wide variety of cellular processes. ALT-associated PML bodies (APBs) are specialized PML bodies found exclusively in telomerase-negative tumors in which telomeres are maintained by recombination-based alternative (ALT) mechanisms. Although it has been suggested that APBs are directly implicated in telomere metabolism of ALT cells, their precise role and structure have remained elusive. Here we show that PML bodies in ALT cells associate with chromosome ends forming small, spatially well-defined clusters, containing on average 2-5 telomeres. Using an innovative approach that gently enlarges PML bodies in living cells while retaining their overall organization, we show that this physical enlargement of APBs spatially resolves the single telomeres in the cluster, but does not perturb the potential of the APB to recruit chromosome extremities. We show that telomere clustering in PML bodies is cell-cycle regulated and that unique telomeres within a cluster associate with recombination proteins. Enlargement of APBs induced the accumulation of telomere-telomere recombination intermediates visible on metaphase spreads and connecting heterologous chromosomes. The strand composition of these recombination intermediates indicated that this recombination is constrained to a narrow time window in the cell cycle following replication. These data provide strong evidence that PML bodies are not only a marker for ALT cells but play a direct role in telomere recombination, both by bringing together chromosome ends and by promoting telomere-telomere interactions between heterologous chromosomes.


Asunto(s)
Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Recombinación Genética , Telómero/genética , Telómero/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ciclo Celular/genética , Ciclo Celular/fisiología , Línea Celular , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Hibridación Fluorescente in Situ , Espacio Intranuclear/metabolismo , Proteína de la Leucemia Promielocítica , Telomerasa/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
9.
J Clin Invest ; 119(5): 1093-108, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19363294

RESUMEN

The identification of biomarkers that distinguish between aggressive and indolent forms of prostate cancer (PCa) is crucial for diagnosis and treatment. In this study, we used cultured cells derived from prostate tissue from patients with PCa to define a molecular mechanism underlying the most aggressive form of PCa that involves the functional activation of eNOS and HIFs in association with estrogen receptor beta (ERbeta). Cells from patients with poor prognosis exhibited a constitutively hypoxic phenotype and increased NO production. Upon estrogen treatment, formation of ERbeta/eNOS, ERbeta/HIF-1alpha, or ERbeta/HIF-2alpha combinatorial complexes led to chromatin remodeling and transcriptional induction of prognostic genes. Tissue microarray analysis, using an independent cohort of patients, established a hierarchical predictive power for these proteins, with expression of eNOS plus ERbeta and nuclear eNOS plus HIF-2alpha being the most relevant indicators of adverse clinical outcome. Genetic or pharmacologic modulation of eNOS expression and activity resulted in reciprocal conversion of the transcriptional signature in cells from patients with bad or good outcome, respectively, highlighting the relevance of eNOS in PCa progression. Our work has considerable clinical relevance, since it may enable the earlier diagnosis of aggressive PCa through routine biopsy assessment of eNOS, ERbeta, and HIF-2alpha expression. Furthermore, proposing eNOS as a therapeutic target fosters innovative therapies for PCa with NO inhibitors, which are employed in preclinical trials in non-oncological diseases.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Receptor beta de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Neoplasias de la Próstata/diagnóstico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biomarcadores , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Citoplasma/metabolismo , Inhibidores Enzimáticos/farmacología , Estradiol/farmacología , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Transportador de Glucosa de Tipo 1/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/genética , Pronóstico , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Elementos de Respuesta/genética , Telomerasa/genética , Telomerasa/metabolismo , Análisis de Matrices Tisulares , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
10.
J Appl Physiol (1985) ; 106(1): 333-7, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19023015

RESUMEN

Hormones and nitric oxide (NO), a free radical, are ancestral molecules, conserved through evolution, that modulate many aspects of the physiology and pathophysiology of living organisms by regulating transcription of genes involved in development, metabolism, and differentiation. Of interest, both estrogen and NO signaling, specifically through the estrogen receptor-alpha (ERalpha) and the endothelial isoform of the nitric oxide synthase (eNOS), have been shown to counteract endothelial senescence through a shared downstream effector, the catalytic subunit of human telomerase (hTERT), a key molecule in the aging process. Since aging is the first and most relevant risk factor in cardiovascular diseases, it is tempting to speculate that hTERT may be at the cross point between the NO and estrogen pathways. The present review will focus on the evolutionary and molecular aspects linking eNOS, ERs, and hTERT in counteracting the process of endothelial cell aging.


Asunto(s)
Senescencia Celular , Endotelio Vascular/enzimología , Estrógenos/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Telomerasa/metabolismo , Endotelio Vascular/patología , Humanos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptores de Estrógenos/metabolismo
11.
Circ Res ; 103(1): 34-42, 2008 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-18519947

RESUMEN

We report that in endothelial cells, the angiogenic effect of 17beta-estradiol (E2) is inhibited by the estrogen receptor (ER) antagonist ICI or the NO synthase (NOS) inhibitor 7-nitroindazole via downregulation of hTERT, the telomerase catalytic subunit, suggesting that E2 and NO are involved in controlling hTERT transcription. Quantitative Real-Time PCR and chromatin immunoprecipitations in E2-treated human umbilical vein endothelial cells, showed recruitment of ERs on the hTERT promoter and concomitant enrichment in histone 3 methylation at Lysine 79, a modification associated with transcription-competent chromatin. Confocal microscopy and re-chromatin immunoprecipitations revealed that on E2 induction, endothelial (e)NOS rapidly localized into the nucleus and associated with ERalpha on the hTERT promoter. Transfections of a constitutively active eNOS mutant (S1177D) strongly induced the hTERT promoter, indicating a direct role of the protein in hTERT transcriptional regulation. Mutation of the estrogen response element in the promoter abolished response to both ERs and active eNOS, demonstrating that the estrogen response element integrity is required for hTERT regulation by these factors. To investigate this novel regulation in a reduced NO environment, pulmonary endothelial cells were isolated from eNOS(-/-) mice and grown with/without E2. In wild-type cells, E2 significantly increased telomerase activity. In eNOS(-/-) cells, basal telomerase activity was rescued by exogenous eNOS or an NO donor, whereas responsiveness to E2 demanded the active protein. In conclusion, we document the novel findings of a combinatorial eNOS/ERalpha complex at the hTERT estrogen response element site and that active eNOS and ligand-activated ERs cooperate in regulating hTERT expression in the endothelium.


Asunto(s)
Núcleo Celular/enzimología , Células Endoteliales/enzimología , Receptor alfa de Estrógeno/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Telomerasa/biosíntesis , Transcripción Genética/fisiología , Venas Umbilicales/enzimología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Sustitución de Aminoácidos , Animales , Bovinos , Células Cultivadas , Metilación de ADN , Células Endoteliales/citología , Inhibidores Enzimáticos/farmacología , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Estrógenos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Indazoles/farmacología , Ratones , Ratones Noqueados , Mutación Missense , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Elementos de Respuesta/fisiología , Telomerasa/genética , Transcripción Genética/efectos de los fármacos , Transfección , Venas Umbilicales/citología
12.
Mol Cancer Res ; 4(2): 79-92, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16513839

RESUMEN

The histopathologic and molecular heterogeneity of prostate cancer and the limited availability of human tumor tissue make unraveling the mechanisms of prostate carcinogenesis a challenging task. Our goal was to develop an ex vivo model that could be reliably used to define a prognostic signature based on gene expression profiling of cell cultures that maintained the tumor phenotype. To this end, we derived epithelial cultures from tissue explanted from 59 patients undergoing radical prostatectomy or cistoprostatectomy because of prostate benign hyperplasia/prostate cancer or bladder carcinoma. Patient selection criteria were absence of hormonal neoadjuvant treatment before surgery and diagnosis of clinically localized disease. Using this unique experimental material, we analyzed expression of 22,500 transcripts on the Affymetrix Human U133A GeneChip platform (Affymetrix, Inc., High Wycombe, United Kingdom). Cultures from normal/hyperplastic tissues with a prevalent luminal phenotype and from normal prostate epithelial tissue with basal phenotype (PrEC) served as controls. We have established a large number of prostate primary cultures highly enriched in the secretory phenotype. From them, we derived an epithelial-restricted transcriptional signature that (a) differentiated normal from tumor cells and (b) clearly separated cancer-derived lines into two distinct groups, which correlated with indolent or aggressive clinical behavior of the disease. Our findings provide (a) a method to expand human primary prostate carcinoma cells with a luminal phenotype, (b) a powerful experimental model to study primary prostate cancer biology, and (c) a novel means to characterize these tumors from a molecular genetic standpoint for prognostic and/or predictive purposes.


Asunto(s)
Biomarcadores de Tumor/genética , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Neoplasias de la Próstata/genética , Anciano , Diferenciación Celular , Células Cultivadas , Células Epiteliales/patología , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Pronóstico , Próstata/metabolismo , Prostatectomía , Hiperplasia Prostática/genética , Hiperplasia Prostática/patología , Neoplasias de la Próstata/cirugía , Células Tumorales Cultivadas
13.
Oncogene ; 24(53): 7893-901, 2005 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-16116482

RESUMEN

In human somatic cells proliferation results in telomere shortening due to the end replication problem and the absence of adequate levels of telomerase activity. The progressive loss of telomeric DNA has been associated with replicative senescence. Maintenance of telomere structure and function is, therefore, an essential requisite for cells that proliferate indefinitely. Human cells that have acquired the immortal phenotype mostly rely on telomerase to compensate for telomere shortening with cell division. However, a certain percentage of immortalized cell lines and human tumors maintain their telomeres by Alternative Lengthening of Telomeres (ALT), a mechanism not fully understood but apparently based on homologous recombination. Here, we report the isolation of an immortal human cell line that is derived from an ALT cell line but maintains telomeres in the absence of key features of ALT and of telomerase. The properties of these cells suggest that the identification of ALT cells may not be reliably based on known ALT markers. This finding is of relevance for discriminating between the mortal and immortal phenotype among telomerase-negative cells in vitro and in vivo, particularly in regard to the development of pharmacological approaches for cancer treatment based on telomerase inhibition.


Asunto(s)
Telomerasa/metabolismo , Telómero/ultraestructura , Células Tumorales Cultivadas , Biomarcadores/análisis , Proliferación Celular , Humanos , Fenotipo , Transfección
14.
J Biol Chem ; 280(15): 14790-8, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15687494

RESUMEN

Telomere dysfunction contributes to reduced cell viability, altered differentiation, and impaired regenerative/proliferative responses. Recent advances indicate that telomerase activity confers a pro-angiogenic phenotype to endothelial cells and their precursors. We have investigated whether telomerase contributes to tissue regeneration following hind limb ischemia and vascular endothelial growth factor 165 (VEGF(165)) treatment. VEGF delivery induced angiogenesis and increased expression of the telomerase reverse transcriptase (TERT) and telomerase activity in skeletal muscles and satellite and endothelial cells. Adenovirus-mediated transfer of wild type TERT but not of a dominant negative mutant, TERTdn, significantly induced capillary but not arteriole formation. However, when co-delivered with VEGF, TERTdn abrogated VEGF-dependent angiogenesis, arteriogenesis, and blood flow increase. This effect was paralleled by in vitro evidence that telomerase inhibition by 3'-azido-3'-deoxythymidine in VEGF-treated endothelial cells strongly reduced capillary density and promoted apoptosis in the absence of serum. Similar results were obtained with adenovirus-mediated expression of TERTdn and AKTdn, both reducing endogenous TERT activity and angiogenesis on Matrigel. Mechanistically, neo-angiogenesis in our system involved: (i) VEGF-dependent activation of telomerase through the nitric oxide pathway and (ii) telomerase-dependent activation of endothelial cell differentiation and protection from apoptosis. Furthermore, detection of TERT in activated satellite cells identified them as VEGF targets during muscle regeneration. Because TERT behaves as an angiogenic factor and a downstream effector of VEGF signaling, telomerase activity appears required for VEGF-dependent remodeling of ischemic tissue at the capillaries and arterioles level.


Asunto(s)
Isquemia , Telomerasa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adenoviridae/genética , Animales , Apoptosis , Ciclo Celular , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Cultivadas , Colágeno/química , Proteínas de Unión al ADN , Combinación de Medicamentos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Extremidades , Terapia Genética/métodos , Vectores Genéticos , Humanos , Etiquetado Corte-Fin in Situ , Laminina/química , Ratones , Microscopía Fluorescente , Músculo Esquelético/metabolismo , Neovascularización Patológica , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Perfusión , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Proteoglicanos/química , Ratas , Regeneración , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transcripción Genética , Transfección , Venas Umbilicales/citología , Regulación hacia Arriba
15.
Cancer Res ; 64(7): 2324-7, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15059879

RESUMEN

Telomere maintenance activity is a hallmark of cancer. In some telomerase-negative tumors, telomeres become lengthened by alternative lengthening of telomeres (ALT), a recombination-mediated DNA replication process in which telomeres use other telomeric DNA as a copy template. Using chromosome orientation fluorescence in situ hybridization, we found that postreplicative exchange events involving a telomere and another TTAGGG-repeat tract occur at remarkably high frequencies in ALT cells (range 28-280/100 metaphases) and rarely or never in non-ALT cells, including cell lines with very long telomeres. Like the ALT phenotype itself, the telomeric exchanges were not suppressed when telomerase was activated in ALT cells. These exchanges are telomere specific because there was no correlation with sister chromatid exchange rates at interstitial locations, and they were not observed in non-ALT Bloom syndrome cells with very high sister chromatid exchange rates.


Asunto(s)
Neoplasias/genética , Telómero/genética , Animales , Línea Celular Tumoral , Humanos , Hibridación Fluorescente in Situ , Ratones , Células 3T3 NIH , Intercambio de Cromátides Hermanas
16.
Oncogene ; 23(6): 1221-8, 2004 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-14716292

RESUMEN

Maintenance of telomeres is essential for chromosome stability. In the absence of telomerase, telomeres shorten with cell division until they approach a stability threshold, at which point cells enter senescence. When senescence-signaling pathways are inactive, further telomere shortening leads to chromosome instability characterized by telomeric fusions and breakage-fusion-bridge (BFB) cycles. Since the distribution of telomere lengths among chromosome extremities is heterogeneous, we wondered about the impact of such variability on the stability of particular chromosome arms. We correlated the initial length of individual telomeres in telomerase-negative-transformed cells with the stability of the corresponding chromosome arms during the precrisis period. We show that arms carrying the shortest telomeres are the first to become unstable and this instability affects the chromosome homologues with shorter telomeres almost exclusively. The analysis of several postcrisis cell populations, which had stabilized their telomeres by re-expressing telomerase, showed that the karyotypic outcome is strongly influenced by the initial telomere length heterogeneity. The timing of telomerase re-expression also seems to play a role in limiting the extent of karyotypic changes, probably by reducing the frequency of telomeric fusions and hence BFB. Since the distribution of telomere lengths within somatic cells is proper to every individual, our results predict that the risk for a particular chromosome arm of becoming unstable early in tumorigenesis will differ between individuals and contribute directly to the heterogeneity of chromosome aberrations found in tumors.


Asunto(s)
Transformación Celular Neoplásica/genética , Aberraciones Cromosómicas , Evolución Molecular , Neoplasias/genética , Telómero/genética , División Celular , Línea Celular , Células Clonales , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Virus 40 de los Simios/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...