Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Oncogenesis ; 4: e176, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26657580

RESUMEN

Here we report that promoter mutations in telomerase (TERT), the most common noncoding mutations in cancer, give rise to monoallelic expression of TERT. Through deep RNA sequencing, we find that TERT activation in human cancer cell lines can occur in either mono- or biallelic manner. Without exception, hotspot TERT promoter mutations lead to the re-expression of only one allele, accounting for approximately half of the observed cases of monoallelic TERT expression. Furthermore, we show that monoallelic TERT expression is highly prevalent in certain tumor types and widespread across a broad spectrum of cancers. Taken together, these observations provide insights into the mechanisms of TERT activation and the ramifications of noncoding mutations in cancer.

2.
Oncogene ; 34(2): 209-16, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-24362534

RESUMEN

Aberrant nuclear factor (NF)-κB activation is frequently observed in human cancers. Genome characterization efforts have identified genetic alterations in multiple components of the NF-κB pathway, some of which have been shown to be essential for cancer initiation and tumor maintenance. Here, using patient tumors and cancer cell lines, we identify the NF-κB regulator, TRAF2 (tumor necrosis factor (TNF) receptor-associated factor 2), as an oncogene that is recurrently amplified and rearranged in 15% of human epithelial cancers. Suppression of TRAF2 in cancer cells harboring TRAF2 copy number gain inhibits proliferation, NF-κB activation, anchorage-independent growth and tumorigenesis. Cancer cells that are dependent on TRAF2 also require NF-κB for survival. The phosphorylation of TRAF2 at serine 11 is essential for the survival of cancer cells harboring TRAF2 amplification. Together, these observations identify TRAF2 as a frequently amplified oncogene.


Asunto(s)
FN-kappa B/genética , FN-kappa B/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Factor 2 Asociado a Receptor de TNF/genética , Factor 2 Asociado a Receptor de TNF/metabolismo , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Xenoinjertos , Humanos , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Desnudos , Oncogenes , Fosforilación , Transducción de Señal
3.
Oncogene ; 34(16): 2061-71, 2015 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-24909179

RESUMEN

Targeting HER2 with antibodies or small molecule inhibitors in HER2-positive breast cancer leads to improved survival, but resistance is a common clinical problem. To uncover novel mechanisms of resistance to anti-HER2 therapy in breast cancer, we performed a kinase open reading frame screen to identify genes that rescue HER2-amplified breast cancer cells from HER2 inhibition or suppression. In addition to multiple members of the MAPK (mitogen-activated protein kinase) and PI3K (phosphoinositide 3-kinase) signaling pathways, we discovered that expression of the survival kinases PRKACA and PIM1 rescued cells from anti-HER2 therapy. Furthermore, we observed elevated PRKACA expression in trastuzumab-resistant breast cancer samples, indicating that this pathway is activated in breast cancers that are clinically resistant to trastuzumab-containing therapy. We found that neither PRKACA nor PIM1 restored MAPK or PI3K activation after lapatinib or trastuzumab treatment, but rather inactivated the pro-apoptotic protein BAD, the BCl-2-associated death promoter, thereby permitting survival signaling through BCL-XL. Pharmacological blockade of BCL-XL/BCL-2 partially abrogated the rescue effects conferred by PRKACA and PIM1, and sensitized cells to lapatinib treatment. These observations suggest that combined targeting of HER2 and the BCL-XL/BCL-2 anti-apoptotic pathway may increase responses to anti-HER2 therapy in breast cancer and decrease the emergence of resistant disease.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/fisiología , Resistencia a Antineoplásicos/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Receptor ErbB-2/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/genética , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Lapatinib , Proteínas Quinasas Activadas por Mitógenos/genética , Sistemas de Lectura Abierta/genética , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Proteínas Proto-Oncogénicas c-pim-1/genética , Trastuzumab , Proteína Letal Asociada a bcl/antagonistas & inhibidores , Proteína Letal Asociada a bcl/metabolismo , Proteína bcl-X/antagonistas & inhibidores
4.
Oncogene ; 33(49): 5637-48, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-24317510

RESUMEN

Lineage-restricted transcription factors (TFs) are frequently mutated or overexpressed in cancer and contribute toward malignant behaviors; however, the molecular bases of their oncogenic properties are largely unknown. As TF activities are difficult to inhibit directly with small molecules, the genes and pathways they regulate might represent more tractable targets for drug therapy. We studied GATA6, a TF gene that is frequently amplified or overexpressed in gastric, esophageal and pancreatic adenocarcinomas. GATA6-overexpressing gastric cancer cell lines cluster in gene expression space, separate from non-overexpressing lines. This expression clustering signifies a shared pathogenic group of genes that GATA6 may regulate through direct cis-element binding. We used chromatin immunoprecipitation and sequencing (ChIP-seq) to identify GATA6-bound genes and considered TF occupancy in relation to genes that respond to GATA6 depletion in cell lines and track with GATA6 mRNA (synexpression groups) in primary gastric cancers. Among other cellular functions, GATA6-occupied genes control apoptosis and govern the M-phase of the cell cycle. Depletion of GATA6 reduced the levels of the latter transcripts and arrested cells in G2 and M phases of the cell cycle. Synexpression in human tumor samples identified likely direct transcriptional targets substantially better than consideration only of transcripts that respond to GATA6 loss in cultured cells. Candidate target genes responded to the loss of GATA6 or its homolog GATA4 and even more to the depletion of both proteins. Many GATA6-dependent genes lacked nearby binding sites but several strongly dependent, synexpressed and GATA6-bound genes encode TFs such as MYC, HES1, RARB and CDX2. Thus, many downstream effects occur indirectly through other TFs and GATA6 activity in gastric cancer is partially redundant with GATA4. This integrative analysis of locus occupancy, gene dependency and synexpression provides a functional signature of GATA6-overexpressing gastric cancers, revealing both limits and new therapeutic directions for a challenging and frequently fatal disease.


Asunto(s)
Factor de Transcripción GATA6/genética , Factor de Transcripción GATA6/fisiología , Regulación Neoplásica de la Expresión Génica , Neoplasias Gástricas/metabolismo , Apoptosis , Sitios de Unión , Ciclo Celular , Línea Celular Tumoral , Linaje de la Célula , Proliferación Celular , Epigénesis Genética , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Perfilación de la Expresión Génica , Histonas/metabolismo , Humanos , ARN Mensajero/genética , Transducción de Señal , Factores de Transcripción/metabolismo
5.
Oncogene ; 31(29): 3397-408, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22105362

RESUMEN

Activating mutations in the RAS family or BRAF frequently occur in many types of human cancers but are rarely detected in breast tumors. However, activation of the RAS-RAF-MEK-ERK MAPK pathway is commonly observed in human breast cancers, suggesting that other genetic alterations lead to activation of this signaling pathway. To identify breast cancer oncogenes that activate the MAPK pathway, we screened a library of human kinases for their ability to induce anchorage-independent growth in a derivative of immortalized human mammary epithelial cells (HMLE). We identified p21-activated kinase 1 (PAK1) as a kinase that permitted HMLE cells to form anchorage-independent colonies. PAK1 is amplified in several human cancer types, including 30--33% of breast tumor samples and cancer cell lines. The kinase activity of PAK1 is necessary for PAK1-induced transformation. Moreover, we show that PAK1 simultaneously activates MAPK and MET signaling; the latter via inhibition of merlin. Disruption of these activities inhibits PAK1-driven anchorage-independent growth. These observations establish PAK1 amplification as an alternative mechanism for MAPK activation in human breast cancer and credential PAK1 as a breast cancer oncogene that coordinately regulates multiple signaling pathways, the cooperation of which leads to malignant transformation.


Asunto(s)
Neoplasias de la Mama/patología , Sistema de Señalización de MAP Quinasas/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Oncogenes , Proteínas Proto-Oncogénicas c-met/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Activación Enzimática/genética , Genoma Humano/genética , Humanos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
6.
Oncogene ; 30(6): 631-41, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21042276

RESUMEN

The IκB Kinase (IKK)-related kinases TBK1 and IKKɛ have essential roles as regulators of innate immunity by modulating interferon and NF-κB signaling. Recent work has also implicated these non-canonical IKKs in malignant transformation. IKKɛ is amplified in ∼30% of breast cancers and transforms cells through the activation of NF-κB. TBK1 participates in RalB-mediated inflammatory responses and cell survival, and is essential for the survival of non-small cell lung cancers driven by oncogenic KRAS. The delineation of target substrates and downstream activities for TBK1 and IKKɛ has begun to define their role(s) in promoting tumorigenesis. In this review, we will highlight the mechanisms by which IKKɛ and TBK1 orchestrate pathways involved in inflammation and cancer.


Asunto(s)
Adenocarcinoma/enzimología , Neoplasias de la Mama/enzimología , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Quinasa I-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Adenocarcinoma/inmunología , Apoptosis/inmunología , Neoplasias de la Mama/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Transformación Celular Neoplásica/metabolismo , Femenino , Humanos , Quinasa I-kappa B/inmunología , Inmunidad Innata , Inflamación/enzimología , Inflamación/inmunología , Interferones/inmunología , FN-kappa B/inmunología , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Transducción de Señal/inmunología , Proteínas ras/inmunología , Proteínas ras/metabolismo
7.
Oncogene ; 27(26): 3754-60, 2008 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-18223679

RESUMEN

The expression level of the telomerase catalytic subunit (telomerase reverse transcriptase, TERT) positively correlates with cell survival after exposure to several lethal stresses. However, whether the protective role of TERT is independent of telomerase activity has not yet been clearly explored. Here, we genetically evaluated the protective roles of both TERT and telomerase activity against cell death induced by staurosporine (STS) and N-methyl-D-aspartic acid (NMDA). First generation (G1) TERT-deficient mouse embryonic fibroblasts (MEFs) displayed an increased sensitivity to STS, while TERT transgenic MEFs were more resistant to STS-induced apoptosis than wild-type. Deletion of the telomerase RNA component (TERC) failed to alter the sensitivity of TERT transgenic MEFs to STS treatment. Similarly, NMDA-induced excitotoxic cell death of primary neurons was suppressed by TERT, but not by TERC both in vitro and in vivo. Specifically, NMDA accelerated death of TERT-deficient mice, while TERT transgenic mice showed enhanced survival when compared with wild-type littermates after administration of NMDA. In addition, the transgenic expression of TERT protected motor neurons from apoptosis induced by sciatic nerve axotomy. These results indicate that telomerase activity is not essential for the protective function of TERT. This telomerase activity-independent TERT function may contribute to cancer development and aging independently of telomere lengthening.


Asunto(s)
Telomerasa/fisiología , Animales , Apoptosis , Calcio/metabolismo , Supervivencia Celular , Humanos , Ratones , Ratones Transgénicos , N-Metilaspartato/toxicidad , Estaurosporina/farmacología , Telómero
8.
Curr Mol Med ; 5(2): 227-31, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15974877

RESUMEN

Accumulating evidence now implicates telomeres and telomerase as critical regulators genomic stability and replicative lifespan in mammalian cells. Disruption of telomere maintenance and/or telomerase expression contributes to the etiology of some degenerative diseases and may participate in the process of aging. Although telomere dysfunction and aberrant telomerase expression clearly play important roles in cancer development, the contribution of telomere biology to cancer is complex and involves both positive and negative influences on tumor development. Indeed, recent work from several laboratories suggests additional roles for telomeres and telomerase in both normal and malignant physiology. Understanding the complexity of telomere biology will provide further insights into chromosome biology in both normal and malignant cells.


Asunto(s)
Senescencia Celular , Inestabilidad Genómica , Telomerasa/metabolismo , Telómero/metabolismo , Humanos
9.
Histol Histopathol ; 18(2): 541-50, 2003 04.
Artículo en Inglés | MEDLINE | ID: mdl-12647805

RESUMEN

We now understand neoplastic transformation to be the consequence of multiple acquired genetic alterations. The combination of these acquired changes confer the various phenotypes that constitute the clinical features of cancer. Although only rare human cancers derive from a viral etiology, the study of DNA tumor viruses that transform rodent and human cells has led to a greater understanding of the molecular events that program the malignant state. In particular, investigation of the viral oncoproteins specified by the Simian Virus 40 Early Region (SV40 ER) has revealed critical host cell pathways, whose perturbation play an essential role in the experimental transformation of mammalian cells. Recent work has re-investigated the roles of two SV40 ER oncoproteins, the large T antigen (LT) and the small t antigen (ST), in human cell transformation. Co-expression of these two oncoproteins, together with the telomerase catalytic subunit, hTERT, and an oncogenic version of the H-Ras oncoprotein, suffices to transform human cells. LT inactivates two key tumor suppressor pathways by binding to the retinoblastoma protein (pRB) and p53. The ability of ST to transform human cells requires interactions with PP2A, an abundant family of serine-threonine phosphatases. Here we review recent developments in our understanding of how these two viral oncoproteins facilitate human cell transformation.


Asunto(s)
Antígenos Transformadores de Poliomavirus/fisiología , Transformación Celular Neoplásica/patología , Animales , Humanos
10.
Cancer Res ; 61(23): 8366-70, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11731409

RESUMEN

Although high frequencies of T lymphocytes specific for certain tumor-associated antigens have been detected in some cancer patients, increasing evidence suggests that these T cells may be functionally defective in vivo and fail to induce meaningful clinical responses. One strategy to overcome this limitation is to target novel antigens that are ignored during the natural antitumor immune response but are nevertheless capable of triggering effector T-cell responses against tumors after optimal presentation by antigen-presenting cells. Here, we show that the telomerase catalytic subunit (hTERT)-a nearly universal tumor antigen identified by epitope deduction rather than from patient immune responses-is immunologically ignored by patients despite progressive tumor burden. Nevertheless, HLA-A2-restricted CTLs against hTERT are equivalently induced ex vivo from patients and healthy individuals and efficiently kill human tumor cell lines and primary tumors. Thus, telomerase-specific T cells from cancer patients are spared functional inactivation because of immunological ignorance. These findings support clinical efforts to target the hTERT as a tumor antigen with broad therapeutic potential.


Asunto(s)
Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Telomerasa/inmunología , Adulto , Anciano , Proteínas de Unión al ADN , Epítopos de Linfocito T/inmunología , Femenino , Antígeno HLA-A2/inmunología , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/inmunología
11.
Cancer Res ; 61(24): 8838-44, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11751406

RESUMEN

The majority of adult human epithelial cancers exhibit evidence of genetic instability, and it is widely believed that the genetic instability manifested by aneuploidy or microsatellite instability plays an essential role in the genesis of these tumors. Indeed, most experimental models of cancer also show evidence of genomic instability. The resulting genetic chaos, which has widespread effects on many genes throughout the genome, confounds attempts to determine the precise cohort of genetic changes that are required for the transformation of normal human cells to a tumorigenic state. Here we show that genetic transformation of human kidney epithelial cells can occur in the absence of extensive aneuploidy, chromosomal translocations, and microsatellite instability. These observations demonstrate that the in vitro oncogenic transformation of human cells can proceed without widespread genomic instability.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias/genética , Línea Celular Transformada , Proteínas de Unión al ADN , Fibroblastos/citología , Fibroblastos/fisiología , Genes Inmediatos-Precoces , Genes ras , Genoma Humano , Humanos , Cariotipificación , Riñón/citología , Riñón/fisiología , Virus 40 de los Simios/genética , Telomerasa/genética
12.
Clin Cancer Res ; 7(11): 3343-8, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11705846

RESUMEN

PURPOSE: We have reported previously that the telomerase catalytic subunit, human telomerase reverse transcriptase (hTERT), is a widely expressed tumor-associated antigen recognized by CTLs. A nine-amino acid peptide derived from hTERT binds strongly to HLA-A2 antigen and elicits CTL responses against a broad panel of hTERT+ tumors (but not hTERT+ hematopoietic progenitor cells). The applicability of hTERT as a potential target for anticancer immunotherapy would be widened by the identification of epitopes restricted to other common HLA alleles, such as HLA-A3 antigen. EXPERIMENTAL DESIGN: Using a method of epitope deduction, HLA-A3-restricted peptide epitopes were screened from hTERT and tested for immunogenicity in a human in vitro T-cell system. RESULTS: The hTERT peptide K973 was used to generate specific CD8+ CTLs from HLA-A3+ cancer patients and healthy individuals. These CTLs lysed hTERT+ tumors from multiple histologies in an MHC-restricted fashion, suggesting that the epitope is naturally processed and presented by tumors. In contrast, highly enriched HLA-A3+ CD34+ peripheral blood progenitor cells or activated T cells were not lysed. CONCLUSION: Given the expression of HLA-A2 and HLA-A3 antigen in the general population, these findings extend the potential applicability of hTERT as a therapeutic target to >60% of all cancer patients. The characterization of hTERT as a polyepitope, polyallelic tumor-associated antigen may provide an approach for circumventing therapy-induced resistance potentially mediated by antigenic- and allelic-loss tumor escape mutants.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígeno HLA-A3/inmunología , Linfocitos T Citotóxicos/inmunología , Telomerasa/inmunología , Secuencia de Aminoácidos , Linfocitos B/enzimología , Linfocitos B/inmunología , Antígenos CD40/análisis , Citotoxicidad Inmunológica , Proteínas de Unión al ADN , Epítopos/química , Epítopos/inmunología , Epítopos/metabolismo , Antígeno HLA-A3/metabolismo , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/inmunología , Humanos , Leucocitos Mononucleares/enzimología , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Unión Proteica , Telomerasa/química , Telomerasa/metabolismo , Células Tumorales Cultivadas
14.
J Clin Invest ; 108(5): 725-32, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11544278

RESUMEN

The immortalization of human cells is a critical step in multistep carcinogenesis. Oral-esophageal carcinomas, a model system to investigate molecular mechanisms underlying squamous carcinogenesis, frequently involve cyclin D1 overexpression and inactivation of the p53 tumor suppressor. Therefore, our goal was to establish the functional role of cyclin D1 overexpression and p53 inactivation in the immortalization of primary human oral squamous epithelial cells (keratinocytes) as an important step toward malignant transformation. Cyclin D1 overexpression alone was found to induce extension of the replicative life span of normal oral keratinocytes, whereas the combination of cyclin D1 overexpression and p53 inactivation led to their immortalization. This study also demonstrates that immortalization of oral keratinocytes can be independent of telomerase activation, involving an alternative pathway of telomere maintenance (ALT).


Asunto(s)
Carcinoma de Células Escamosas/etiología , Transformación Celular Neoplásica , Ciclina D1/genética , Genes p53 , Queratinocitos/fisiología , Neoplasias de la Boca/etiología , Carcinoma de Células Escamosas/enzimología , Ciclo Celular , División Celular , Línea Celular , Ciclina D1/metabolismo , Humanos , Queratinocitos/citología , Queratinocitos/enzimología , Boca/citología , Neoplasias de la Boca/enzimología , Mutación , Telomerasa/fisiología , Transducción Genética
15.
Ann Med ; 33(2): 123-9, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11327115

RESUMEN

The maintenance of specialized nucleoprotein structures termed telomeres is essential for chromosome stability. Without new synthesis of telomeres at chromosome ends the chromosomes shorten with progressive cell division, eventually triggering either replicative senescence or apoptosis when telomere length becomes critically short. The regulation of telomerase activity in human cells plays a significant role in the development of cancer. Telomerase is tightly repressed in the vast majority of normal human somatic cells but becomes activated during cellular immortalization and in cancers. While the mechanisms for telomerase activation in cancers have not been fully defined, they include telomerase catalytic subunit gene (hTERT) amplification and trans-activation of the hTERT promoter by the myc oncogene product. Ectopic expression of hTERT is sufficient to restore telomerase activity in cells that lack the enzyme and can immortalize many cell types. Understanding telomerase biology will eventually lead to several clinically relevant telomerase-based therapies. These applications include inhibiting or targeting telomerase as a novel antineoplastic strategy and using cells immortalized by telomerase for therapeutic applications.


Asunto(s)
Senescencia Celular/fisiología , Neoplasias/fisiopatología , Telomerasa/fisiología , Telómero/fisiología , Dominio Catalítico/fisiología , Senescencia Celular/genética , Proteínas de Unión al ADN , Genes myc/genética , Genes myc/fisiología , Humanos , Neoplasias/genética , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética , Transcripción Genética
16.
Genes Dev ; 15(1): 50-65, 2001 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11156605

RESUMEN

A number of genetic mutations have been identified in human breast cancers, yet the specific combinations of mutations required in concert to form breast carcinoma cells remain unknown. One approach to identifying the genetic and biochemical alterations required for this process involves the transformation of primary human mammary epithelial cells (HMECs) to carcinoma cells through the introduction of specific genes. Here we show that introduction of three genes encoding the SV40 large-T antigen, the telomerase catalytic subunit, and an H-Ras oncoprotein into primary HMECs results in cells that form tumors when transplanted subcutaneously or into the mammary glands of immunocompromised mice. The tumorigenicity of these transformed cells was dependent on the level of ras oncogene expression. Interestingly, transformation of HMECs but not two other human cell types was associated with amplifications of the c-myc oncogene, which occurred during the in vitro growth of the cells. Tumors derived from the transformed HMECs were poorly differentiated carcinomas that infiltrated through adjacent tissue. When these cells were injected subcutaneously, tumors formed in only half of the injections and with an average latency of 7.5 weeks. Mixing the epithelial tumor cells with Matrigel or primary human mammary fibroblasts substantially increased the efficiency of tumor formation and decreased the latency of tumor formation, demonstrating a significant influence of the stromal microenvironment on tumorigenicity. Thus, these observations establish an experimental system for elucidating both the genetic and cell biological requirements for the development of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Mama/citología , Transformación Celular Neoplásica , Células Epiteliales/citología , Genes RAG-1 , Proteínas de Homeodominio/metabolismo , Animales , Antígenos Transformadores de Poliomavirus/genética , Neoplasias de la Mama/patología , División Celular , Femenino , Genes ras , Proteínas de Homeodominio/genética , Humanos , Síndromes de Inmunodeficiencia/genética , Hibridación Fluorescente in Situ , Cariotipificación , Ratones , Ratones Noqueados , Telomerasa/genética , Trasplante Heterólogo
17.
Curr Opin Cell Biol ; 12(6): 705-9, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11063935

RESUMEN

Senescence is now understood to be the final phenotypic state adopted by a cell in response to several distinct cell physiological processes, including proliferation, oncogene activation and oxygen free radical toxicity. The role of telomere maintenance in immortalization and the roles of p16(INK4A), p19(ARF), p53 and other genes in senescence are being further elucidated. Significant progress continues to be made in our understanding of cellular senescence and immortalization.


Asunto(s)
Senescencia Celular/genética , Animales , División Celular/genética , Regulación de la Expresión Génica , Genes p16/genética , Humanos , Ratones , Oncogenes/genética , Estrés Oxidativo/genética , Proteínas/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética , Telómero/genética , Telómero/metabolismo , Proteína p14ARF Supresora de Tumor
18.
Mol Cell Biol ; 20(4): 1436-47, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10648628

RESUMEN

Normal human cells exhibit a limited replicative life span in culture, eventually arresting growth by a process termed senescence. Progressive telomere shortening appears to trigger senescence in normal human fibroblasts and retinal pigment epithelial cells, as ectopic expression of the telomerase catalytic subunit, hTERT, immortalizes these cell types directly. Telomerase expression alone is insufficient to enable certain other cell types to evade senescence, however. Such cells, including keratinocytes and mammary epithelial cells, appear to require loss of the pRB/p16(INK4a) cell cycle control mechanism in addition to hTERT expression to achieve immortality. To investigate the relationships among telomerase activity, cell cycle control, senescence, and differentiation, we expressed hTERT in two epithelial cell types, keratinocytes and mesothelial cells, and determined the effect on proliferation potential and on the function of cell-type-specific growth control and differentiation systems. Ectopic hTERT expression immortalized normal mesothelial cells and a premalignant, p16(INK4a)-negative keratinocyte line. In contrast, when four keratinocyte strains cultured from normal tissue were transduced to express hTERT, they were incompletely rescued from senescence. After reaching the population doubling limit of their parent cell strains, hTERT(+) keratinocytes entered a slow growth phase of indefinite length, from which rare, rapidly dividing immortal cells emerged. These immortal cell lines frequently had sustained deletions of the CDK2NA/INK4A locus or otherwise were deficient in p16(INK4a) expression. They nevertheless typically retained other keratinocyte growth controls and differentiated normally in culture and in xenografts. Thus, keratinocyte replicative potential is limited by a p16(INK4a)-dependent mechanism, the activation of which can occur independent of telomere length. Abrogation of this mechanism together with telomerase expression immortalizes keratinocytes without affecting other major growth control or differentiation systems.


Asunto(s)
Senescencia Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , ARN , Telomerasa/metabolismo , Diferenciación Celular , División Celular , Línea Celular , Transformación Celular Neoplásica , Senescencia Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas de Unión al ADN , Células Epiteliales/citología , Células Epiteliales/metabolismo , Eliminación de Gen , Expresión Génica , Genes p53 , Prueba de Complementación Genética , Humanos , Mutación , Telomerasa/genética
19.
Nat Med ; 5(10): 1164-70, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10502820

RESUMEN

Telomerase is a ribonucleoprotein enzyme that maintains the protective structures at the ends of eukaryotic chromosomes, called telomeres. In most human somatic cells, telomerase expression is repressed, and telomeres shorten progressively with each cell division. In contrast, most human tumors express telomerase, resulting in stabilized telomere length. These observations indicate that telomere maintenance is essential to the proliferation of tumor cells. We show here that expression of a mutant catalytic subunit of human telomerase results in complete inhibition of telomerase activity, reduction in telomere length and death of tumor cells. Moreover, expression of this mutant telomerase eliminated tumorigenicity in vivo. These observations demonstrate that disruption of telomere maintenance limits cellular lifespan in human cancer cells, thus validating human telomerase reverse transcriptase as an important target for the development of anti-neoplastic therapies.


Asunto(s)
Mutación , Neoplasias Experimentales/prevención & control , ARN , Telomerasa/antagonistas & inhibidores , Telomerasa/genética , Apoptosis , Neoplasias de la Mama , Dominio Catalítico/genética , División Celular , Neoplasias del Colon , Proteínas de Unión al ADN , Diseño de Fármacos , Femenino , Vectores Genéticos , Humanos , Neoplasias Experimentales/enzimología , Neoplasias Ováricas , Retroviridae/genética , Inhibidores de la Transcriptasa Inversa , Telómero/metabolismo , Células Tumorales Cultivadas
20.
Nature ; 400(6743): 464-8, 1999 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-10440377

RESUMEN

During malignant transformation, cancer cells acquire genetic mutations that override the normal mechanisms controlling cellular proliferation. Primary rodent cells are efficiently converted into tumorigenic cells by the coexpression of cooperating oncogenes. However, similar experiments with human cells have consistently failed to yield tumorigenic transformants, indicating a fundamental difference in the biology of human and rodent cells. The few reported successes in the creation of human tumour cells have depended on the use of chemical or physical agents to achieve immortalization, the selection of rare, spontaneously arising immortalized cells, or the use of an entire viral genome. We show here that the ectopic expression of the telomerase catalytic subunit (hTERT) in combination with two oncogenes (the simian virus 40 large-T oncoprotein and an oncogenic allele of H-ras) results in direct tumorigenic conversion of normal human epithelial and fibroblast cells. These results demonstrate that disruption of the intracellular pathways regulated by large-T, oncogenic ras and telomerase suffices to create a human tumor cell.


Asunto(s)
Antígenos Transformadores de Poliomavirus/fisiología , Transformación Celular Neoplásica , ARN , Telomerasa/fisiología , Animales , Adhesión Celular , División Celular , Línea Celular , Transformación Celular Neoplásica/genética , Células Cultivadas , Proteínas de Unión al ADN , Células Epiteliales , Fibroblastos , Genes ras , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Telomerasa/genética , Telómero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA