Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Oncogene ; 31(4): 419-31, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-21706059

RESUMEN

Trask/CDCP1 is a transmembrane glycoprotein widely expressed in epithelial tissues whose functions are just beginning to be understood, but include a role as an anti-adhesive effector of Src kinases. Early studies looking at RNA transcript levels seemed to suggest overexpression in some cancers, but immunostaining studies are now providing more accurate analyses of its expression. In an immuno-histochemical survey of human cancer specimens, we find that Trask expression is retained, reduced or sometimes lost in some tumors compared with their normal epithelial tissue counterparts. A survey of human cancer cell lines also show a similar wide variation in the expression of Trask, including some cell types with the loss of Trask expression, and additional cell types that have lost the physiological detachment-induced phosphorylation of Trask. Three experimental models were established to interrogate the role of Trask in tumor progression, including two gain-of-function models with tet-inducible expression of Trask in tumor cells lacking Trask expression, and one loss-of-function model to suppress Trask expression in tumor cells with abundant Trask expression. The induction of Trask expression and phosphorylation in MCF-7 cells and in 3T3v-src cells was associated with a reduction in tumor metastases while the shRNA-induced knockdown of Trask in L3.6pl cancer cells was associated with increased tumor metastases. The results from these three models are consistent with a tumor-suppressing role for Trask. These data identify Trask as one of several potential candidates for functionally relevant tumor suppressors on the 3p21.3 region of the genome frequently lost in human cancers.


Asunto(s)
Antígenos CD/fisiología , Antígenos de Neoplasias/fisiología , Moléculas de Adhesión Celular/fisiología , Glicoproteínas de Membrana/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Línea Celular Tumoral , Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Humanos , Ratones , Células 3T3 NIH , Fosforilación
2.
Oncogene ; 29(40): 5500-10, 2010 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-20661224

RESUMEN

Understanding the mechanisms underlying ErbB3 overexpression in breast cancer will facilitate the rational design of therapies to disrupt ErbB2-ErbB3 oncogenic function. Although ErbB3 overexpression is frequently observed in breast cancer, the factors mediating its aberrant expression are poorly understood. In particular, the ErbB3 gene is not significantly amplified, raising the question as to how ErbB3 overexpression is achieved. In this study we showed that the ZNF217 transcription factor, amplified at 20q13 in ∼20% of breast tumors, regulates ErbB3 expression. Analysis of a panel of human breast cancer cell lines (n = 50) and primary human breast tumors (n = 15) showed a strong positive correlation between ZNF217 and ErbB3 expression. Ectopic expression of ZNF217 in human mammary epithelial cells induced ErbB3 expression, whereas ZNF217 silencing in breast cancer cells resulted in decreased ErbB3 expression. Although ZNF217 has previously been linked with transcriptional repression because of its close association with C-terminal-binding protein (CtBP)1/2 repressor complexes, our results show that ZNF217 also activates gene expression. We showed that ZNF217 recruitment to the ErbB3 promoter is CtBP1/2-independent and that ZNF217 and CtBP1/2 have opposite roles in regulating ErbB3 expression. In addition, we identify ErbB3 as one of the mechanisms by which ZNF217 augments PI-3K/Akt signaling.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 20/genética , Regulación Neoplásica de la Expresión Génica/genética , Receptor ErbB-3/genética , Transactivadores/genética , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Femenino , Expresión Génica , Genes erbB/genética , Humanos , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Oncogenes , Regiones Promotoras Genéticas , Receptor ErbB-3/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transactivadores/metabolismo
3.
J Mammary Gland Biol Neoplasia ; 6(2): 223-34, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11501582

RESUMEN

Acquisition of immortality may be an early and crucial step in malignant progression. We hypothesize that acquisition of unlimited growth potential in individual human mammary epithelial cells (HMEC) requires inactivation of several distinct negative growth constraints as well as reactivation of a mechanism to maintain telomeres on chromosomes. Some of the heritable changes that occur during HMEC immortalization, i.e., loss of expression of cyclin dependent kinase inhibitors p16INK4a and p57KIP2, loss of TGFbeta-mediated growth inhibition, and derepression of telomerase, appear to occur without identifiable mutations in the genes and pathways involved. The absence of mutations, combined with the fact that the changes are often incremental over several cell generations even in clonal populations indicates that some changes associated with immortalization can be epigenetic. We have used the term "conversion" to describe the gradual epigenetic process in chemical carcinogen-immortalized HMEC that leads to activation of telomerase, stabilization of telomere length, and ability to grow uniformly well in the presence or absence of TGFbeta. Characterization of the epigenetic mechanisms involved in immortalization may uncover additional factors that drive tumor progression, and that may be responsive to novel forms of intervention.


Asunto(s)
Mama/citología , Células Epiteliales/fisiología , División Celular/genética , Línea Celular Transformada , Senescencia Celular/genética , Femenino , Humanos , Telomerasa/metabolismo , Telómero/genética
4.
Proc Natl Acad Sci U S A ; 98(8): 4498-503, 2001 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-11287649

RESUMEN

Failures to arrest growth in response to senescence or transforming growth factor beta (TGF-beta) are key derangements associated with carcinoma progression. We report that activation of telomerase activity may overcome both inhibitory pathways. Ectopic expression of the human telomerase catalytic subunit, hTERT, in cultured human mammary epithelial cells (HMEC) lacking both telomerase activity and p16(INK4A) resulted in gaining the ability to maintain indefinite growth in the absence and presence of TGF-beta. The ability to maintain growth in TGF-beta was independent of telomere length and required catalytically active telomerase capable of telomere maintenance in vivo. The capacity of ectopic hTERT to induce TGF-beta resistance may explain our previously described gain of TGF-beta resistance after reactivation of endogenous telomerase activity in rare carcinogen-treated HMEC. In those HMEC that overcame senescence, both telomerase activity and TGF-beta resistance were acquired gradually during a process we have termed conversion. This effect of hTERT may model a key change occurring during in vivo human breast carcinogenesis.


Asunto(s)
Mama/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , ARN , Telomerasa/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Western Blotting , Mama/citología , Dominio Catalítico , División Celular/efectos de los fármacos , Línea Celular Transformada , Proteínas de Unión al ADN , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Humanos , Inmunohistoquímica , Telomerasa/química , Telómero
5.
J Histochem Cytochem ; 49(5): 673-4, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11304810

RESUMEN

Abnormal expression of tyrosine kinase (TK) genes is common in tumors, in which it is believed to alter cell growth and response to external stimuli such as growth factors and hormones. Although the etiology and pathogenesis of carcinomas of the thyroid or breast remain unclear, there is evidence that the expression of TK genes, such as receptor tyrosine kinases, or mitogen-activated protein kinases, is dysregulated in these tumors, and that overexpression of particular TK genes due to gene amplification, changes in gene regulation, or structural alterations leads to oncogenic transformation of epithelial cells. We developed a rapid scheme to measure semiquantitatively the expression levels of 50-100 TK genes. Our assay is based on RT-PCR with mixed based primers that anneal to conserved regions in the catalytic domain of TK genes to generate gene-specific fragments. PCR products are then labeled by random priming and hybridized to DNA microarrays carrying known TK gene targets. Inclusion of differently labeled fragments from reference or normal cells allows identification of TK genes that show altered expression levels during malignant transformation or tumor progression. Examples demonstrate how this innovative assay might help to define new markers for tumor progression and potential targets for disease intervention. (J Histochem Cytochem 49:673-674, 2001)


Asunto(s)
Neoplasias/metabolismo , Proteínas Tirosina Quinasas/genética , Transducción de Señal , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Proteínas Tirosina Quinasas/metabolismo , Neoplasias de la Tiroides/metabolismo , Células Tumorales Cultivadas
6.
Cancer Res ; 61(4): 1250-4, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11245413

RESUMEN

The functional consequences of overexpression of a candidate oncogene on chromosome 20q13.2, ZNF217, were examined by transducing the gene into finite life span human mammary epithelial cells (HMECs). In four independent experiments, ZNF217-transduced cultures gave rise to immortalized cells. HMECs that overcame senescence initially exhibited heterogeneous growth and continued telomere erosion, followed by increasing telomerase activity, stabilization of telomere length, and resistance to transforming growth factor beta growth inhibition. The incremental changes in telomerase activity and growth that occurred in ZNF217-transduced cultures after they overcame senescence were similar to the conversion pattern we have described previously in rare HMEC lines immortalized after exposure to a chemical carcinogen. Aberrant expression of ZNF217 may be selected for during breast cancer progression because it allows breast cells to overcome senescence and attain immortality.


Asunto(s)
Neoplasias de la Mama/genética , Mama/citología , Transformación Celular Neoplásica/genética , Amplificación de Genes , Transactivadores/genética , Mama/metabolismo , Mama/fisiología , Neoplasias de la Mama/patología , Células Cultivadas , Senescencia Celular/genética , Femenino , Humanos , Telomerasa/metabolismo , Telómero/genética , Telómero/metabolismo , Transducción Genética
7.
Carcinogenesis ; 21(5): 871-9, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10783306

RESUMEN

NF-kappaB/Rel is a family of transcription factors which are expressed in all cells; however, in most non-B cells, they are sequestered in the cytoplasm in inactive complexes with specific inhibitory proteins, termed IkappaBs. We have recently shown that NF-kappaB/Rel factors are aberrantly activated in human breast cancer and rodent mammary tumors, and function to promote tumor cell survival and proliferation. Here, we have examined the time-course of induction of NF-kappaB/Rel factors upon carcinogen treatment of female Sprague-Dawley (S-D) rats in vivo and in human mammary epithelial cells (HMECs) in culture. We observed that NF-kappaB/Rel activation is an early event, occurring prior to malignant transformation. In S-D rats, increased NF-kappaB/Rel binding was detected in nuclear extracts of mammary glands from 40% of animals 3 weeks post-treatment with 15 mg/kg 7,12-dimethylbenz[a]anthracene (DMBA); this is prior to formation of tumors which normally begin to be detected after 7-9 weeks. In non-tumorigenic MCF-10F cells, in vitro malignant transformation upon treatment with either DMBA or benzo[a]pyrene (B[a]P) resulted in a 4- to 12-fold increase in activity of classical NF-kappaB (p65/p50). NF-kappaB induction was corrrelated with a decrease in the stability of the NF-kappaB-specific inhibitory protein IkappaB-alpha. Ectopic expression of the transactivating p65 subunit of NF-kappaB in MCF-10F cells induced the c-myc oncogene promoter, which is driven by two NF-kappaB elements, and endogenous c-Myc levels. Furthermore, reduction mammoplasty-derived HMECs, immortalized following B[a]P exposure, showed dysregulated induction of classical NF-kappaB prior to malignant transformation. Together these findings suggest that activation of NF-kappaB plays an early, critical role in the carcinogen-driven transformation of mammary glands.


Asunto(s)
Transformación Celular Neoplásica , Neoplasias Mamarias Experimentales/metabolismo , FN-kappa B/metabolismo , Animales , Secuencia de Bases , Carcinógenos , Femenino , Genes myc , Humanos , Neoplasias Mamarias Experimentales/patología , Datos de Secuencia Molecular , Fenotipo , Ratas , Ratas Sprague-Dawley , Células Tumorales Cultivadas
8.
J Mammary Gland Biol Neoplasia ; 5(4): 365-78, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14973382

RESUMEN

Human pre-malignant breast diseases, particularly ductal carcinoma in situ (DCIS) already display several of the aberrant phenotypes found in primary breast cancers, including chromosomal abnormalities, telomerase activity, inactivation of the p53 gene, and overexpression of some oncogenes. Efforts to model early breast carcinogenesis in human cell cultures have largely involved studies of in vitro transformation of normal finite lifespan human mammary epithelial cells (HMEC) to immortality and malignancy. We present a model of HMEC immortal transformation consistent with the known in vivo data. This model includes a recently described, presumably epigenetic process, termed conversion, which occurs in cells that have overcome stringent replicative senescence and are thus able to maintain proliferation with critically short telomeres. The conversion process involves reactivation of telomerase activity, and acquisition of good uniform growth in the absence and presence of TGFbeta. We propose that overcoming the proliferative constraints set by senescence, and undergoing conversion, represent key rate-limiting steps in human breast carcinogenesis, and occur during early stage breast cancer progression.


Asunto(s)
Neoplasias de la Mama/genética , Células Epiteliales/citología , Glándulas Mamarias Humanas/citología , Neoplasias de la Mama/patología , Transformación Celular Neoplásica , Células Cultivadas , Senescencia Celular , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Humanos , Modelos Biológicos , Fenotipo , Telomerasa/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta/metabolismo , Proteína p53 Supresora de Tumor/fisiología
9.
Cancer Res ; 59(20): 5112-8, 1999 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-10537284

RESUMEN

We have uncovered a novel role for the cyclin-dependent kinase inhibitor, p57KIP2, during the immortalization of cultured human mammary epithelial cells (HMECs). HMECs immortalized after chemical carcinogen exposure initially expressed little or no telomerase activity, and their telomeres continued to shorten with passage. Cell populations whose mean terminal restriction fragment (TRF) length declined to < or = 3 kb exhibited slow heterogeneous growth and contained many nonproliferative cells. These conditionally immortal HMEC cultures accumulated large quantities of p57 protein. With continued passage, the conditionally immortal cell populations very gradually converted to a fully immortal phenotype of good uniform growth, expression of high levels of telomerase activity, and stabilization of telomere length. The fully immortal HMECs that grew well did not accumulate p57 in G0 or during the cell cycle. DNA and RNA analysis of mass populations and individual subclones of conditionally immortal HMEC line 184A1 showed that continued growth of conditionally immortal cells with critically short telomeres was repeatedly accompanied by loss of the expressed p57 allele and transient expression of the allele imprinted previously. Conditionally immortal 184A1 with mean TRF > 3 kb, infected with retroviruses containing the p57 gene, exhibited premature slow heterogeneous growth. Conversely, exogenous expression of human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase, in 184A1 with mean TRF > 3 kb prevented both the slow heterogeneous growth phase and accumulation of p57 in cycling populations. These data indicate that in HMECs that have overcome replicative senescence, p57 may provide an additional barrier against indefinite proliferation. Overcoming p57-mediated growth inhibition in these cells may be crucial for acquisition of the unlimited growth potential thought to be critical for malignant progression.


Asunto(s)
Mama/química , Mama/patología , Transformación Celular Neoplásica , Inhibidores Enzimáticos/análisis , Pérdida de Heterocigocidad , Proteínas Nucleares/análisis , ARN , Alelos , Células Cultivadas , Inhibidor p57 de las Quinasas Dependientes de la Ciclina , Proteínas de Unión al ADN , Células Epiteliales/química , Células Epiteliales/patología , Femenino , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , ARN Mensajero/análisis , Telomerasa/genética , Telómero
10.
Oncogene ; 18(13): 2169-80, 1999 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10327063

RESUMEN

Our recent studies on the process of immortalization of cultured human mammary epithelial cells (HMEC) have uncovered a previously undescribed, apparently epigenetic step, termed conversion. When first isolated, clonally derived HMEC lines of indefinite lifespan showed little or no telomerase activity or ability to maintain growth in the presence of TGFbeta. Cell populations whose mean terminal restriction fragment length had declined to <3 kb also exhibited slow heterogeneous growth, and contained many non-proliferative cells. With continued passage, these conditionally immortal cell populations very gradually converted to a fully immortal phenotype of good growth+/-TGFbeta, expression of high levels of telomerase activity, and stabilization of telomere length. We now show that exposure of the early passage conditionally immortal 184A1 HMEC line to the viral oncogenes human papillomavirus type 16 (HPV16)-E6, -E7, or SV40T, results in either immediate (E6) or rapid (E7; SV40T) conversion of these telomerase negative, TGFbeta sensitive conditionally immortal cells to the fully immortal phenotype. Unlike conditional immortal 184A1, the HPV16-E7 and SV40T exposed cells were able to maintain growth in TGFbeta prior to expression of high levels of telomerase activity. A mutated HPV16-E6 oncogene, unable to inactivate p53, was still capable of rapidly converting conditional immortal 184A1. Our studies provide further evidence that the transforming potential of these viral oncogenes may involve activities beyond their inactivation of p53 and pRB functions. These additional activities may greatly accelerate a step in HMEC immortal transformation, conversion, that would be rate-limiting in the absence of viral oncogene exposure.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Mama/citología , Transformación Celular Viral/genética , Proteínas Oncogénicas Virales/genética , Oncogenes , Papillomaviridae/genética , Proteínas Represoras , Virus 40 de los Simios/genética , Antígenos Transformadores de Poliomavirus/fisiología , División Celular , Supervivencia Celular , Células Cultivadas , Inducción Enzimática , Células Epiteliales/citología , Femenino , Regulación Viral de la Expresión Génica , Humanos , Proteínas Oncogénicas Virales/fisiología , Proteínas E7 de Papillomavirus , Fenotipo , Polimorfismo de Longitud del Fragmento de Restricción , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/fisiología , Proteínas Recombinantes/farmacología , Proteína de Retinoblastoma/antagonistas & inhibidores , Telomerasa/análisis , Telomerasa/biosíntesis , Telómero/química , Transfección , Factor de Crecimiento Transformador beta/farmacología , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
11.
Proc Natl Acad Sci U S A ; 95(15): 8703-8, 1998 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-9671742

RESUMEN

We report here the molecular cloning of an approximately 1-Mb region of recurrent amplification at 20q13.2 in breast cancer and other tumors and the delineation of a 260-kb common region of amplification. Analysis of the 1-Mb region produced evidence for five genes, ZNF217, ZNF218, and NABC1, PIC1L (PIC1-like), CYP24, and a pseudogene CRP (Cyclophillin Related Pseudogene). ZNF217 and NABC1 emerged as strong candidate oncogenes and were characterized in detail. NABC1 is predicted to encode a 585-aa protein of unknown function and is overexpressed in most but not all breast cancer cell lines in which it was amplified. ZNF217 is centrally located in the 260-kb common region of amplification, transcribed in multiple normal tissues, and overexpressed in all cell lines and tumors in which it is amplified and in two in which it is not. ZNF217 is predicted to encode alternately spliced, Kruppel-like transcription factors of 1,062 and 1,108 aa, each having a DNA-binding domain (eight C2H2 zinc fingers) and a proline-rich transcription activation domain.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 20 , Amplificación de Genes , Proteínas de Neoplasias/genética , Transactivadores/genética , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Cromosomas Artificiales de Levadura , Clonación Molecular , Cartilla de ADN , Exones , Humanos , Hibridación Fluorescente in Situ , Intrones , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Transactivadores/química , Transcripción Genética , Células Tumorales Cultivadas , Dedos de Zinc/genética
12.
Mol Biol Cell ; 8(12): 2391-405, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9398663

RESUMEN

Examination of the process of immortal transformation in early passages of two human mammary epithelial cell (HMEC) lines suggests the involvement of an epigenetic step. These lines, 184A1 and 184B5, arose after in vitro exposure of finite lifespan 184 HMEC to a chemical carcinogen, and both are clonally derived. Although early-passage mass cultures of 184A1 and 184B5 maintained continuous slow growth, most individual cells lost proliferative ability. Uniform good growth did not occur until 20-30 passages after the lines first appeared. Early-passage cultures expressed little or no telomerase activity and telomeres continued to shorten with increasing passage. Telomerase activity was first detected when the telomeres became critically short, and activity levels gradually increased thereafter. Early-passage cultures had little or no ability to maintain growth in transforming growth factor-beta (TGFbeta); however, both mass cultures and clonal isolates showed a very gradual increase in the number of cells displaying progressively increased ability to maintain growth in TGFbeta. A strong correlation between capacity to maintain growth in the presence of TGFbeta and expression of telomerase activity was observed. We have used the term "conversion" to describe this process of gradual acquisition of increased growth capacity in the absence or presence of TGFbeta and reactivation of telomerase. We speculate that the development of extremely short telomeres may result in gradual, epigenetic-based changes in gene expression. Understanding the underlying mechanisms of HMEC conversion in vitro may provide new insight into the process of carcinogenic progression in vivo and offer novel modes for therapeutic intervention.


Asunto(s)
Mama/citología , Mama/enzimología , Células Epiteliales/citología , Células Epiteliales/enzimología , Telómero/metabolismo , Adulto , Mama/efectos de los fármacos , Mama/metabolismo , Carcinógenos/farmacología , División Celular/efectos de los fármacos , Línea Celular Transformada , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Humanos , Fenotipo , Telomerasa/metabolismo , Telómero/genética , Factores de Tiempo , Factor de Crecimiento Transformador beta/farmacología
13.
Mol Cell Biol ; 17(5): 2458-67, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9111314

RESUMEN

The effects of transforming growth factor beta (TGF-beta) were studied in closely related human mammary epithelial cells (HMEC), both finite-life-span 184 cells and immortal derivatives, 184A1S, and 184A1L5R, which differ in their cell cycle responses to TGF-beta but express type I and type II TGF-beta receptors and retain TGF-beta induction of extracellular matrix. The arrest-resistant phenotype was not due to loss of cyclin-dependent kinase (cdk) inhibitors. TGF-beta was shown to regulate p15INK4B expression at at least two levels: mRNA accumulation and protein stability. In TGF-beta-arrested HMEC, there was not only an increase in p15 mRNA but also a major increase in p5INK4B protein stability. As cdk4- and cdk6-associated p15INK4B increased during TGF-beta arrest of sensitive cells, there was a loss of cyclin D1, p21Cip1, and p27Kip1 from these kinase complexes, and cyclin E-cdk2-associated p27Kip1 increased. In HMEC, p15INK4B complexes did not contain detectable cyclin. p15INK4B from both sensitive and resistant cells could displace in vitro cyclin D1, p21Cip1, and p27Kip1 from cdk4 isolated from sensitive cells. Cyclin D1 could not be displaced from cdk4 in the resistant 184A1L5R cell lysates. Thus, in TGF-beta arrest, p15INK4B may displace already associated cyclin D1 from cdks and prevent new cyclin D1-cdk complexes from forming. Furthermore, p27Kip1 binding shifts from cdk4 to cyclin E-cdk2 during TGF-beta-mediated arrest. The importance of posttranslational regulation of p15INK4B by TGF-beta is underlined by the observation that in TGF-beta-resistant 184A1L5R, although the p15 transcript increased, p15INK4B protein was not stabilized and did not accumulate, and cyclin D1-cdk association and kinase activation were not inhibited.


Asunto(s)
Mama/metabolismo , Quinasas CDC2-CDC28 , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Inhibidores Enzimáticos/farmacología , Genes Supresores de Tumor , Proteínas Oncogénicas/metabolismo , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor , Ciclina D1 , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Inhibidor p15 de las Quinasas Dependientes de la Ciclina , Femenino , Citometría de Flujo , Humanos , Sustancias Macromoleculares , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Fase de Descanso del Ciclo Celular
14.
J Biol Chem ; 272(15): 10254-9, 1997 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-9092575

RESUMEN

Expression of the complex gene encoding multiple isoforms of structural protein 4.1 is regulated by alternative pre-mRNA splicing. During erythropoiesis, developmental stage-specific inclusion of exon 16 generates protein 4.1 isoforms having a fully functional spectrin-actin binding domain. Here we show that human mammary epithelial cells (HMEC), coincident with the dramatic morphological changes induced by altered culture conditions, exhibit a novel pre-mRNA splicing switch involving a new exon (exon 17B, 450 nucleotides) in the COOH-terminal coding region. 4.1 RNA expressed in proliferating HMEC adherent to culture dishes mostly excluded exon 17B, whereas 4.1 transcripts processed in nondividing suspension cultures of HMEC strongly included this exon. This pre-mRNA splicing switch was reversible: cells transferred from poly(2-hydroxyethyl methacrylate) back to plastic resumed cell division and down-regulated exon 17B expression. More detailed studies revealed complex tissue-specific alternative splicing of exon 17B and another new exon 17A (51 nucleotides). These results predict the existence of multiple 4.1 protein isoforms with diverse COOH termini. Moreover, they strongly suggest that regulation of gene expression during differentiation of epithelial cells is mediated not only by transcriptional mechanisms, but also by post-transcriptional processes such as alternative pre-mRNA splicing.


Asunto(s)
Empalme Alternativo , Mama/citología , Proteínas del Citoesqueleto , Proteínas de la Membrana/genética , Neuropéptidos , Precursores del ARN/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Tamaño de la Célula , Mapeo Cromosómico , Células Epiteliales , Exones , Femenino , Humanos , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular
15.
J Cell Sci ; 110 ( Pt 22): 2861-71, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9427294

RESUMEN

Lactoferrin is a secreted iron binding protein which is expressed during normal functional development of mammary epithelium. Murine mammary epithelial cell lines competent for milk protein expression were used to identify microenvironmental factors that regulate lactoferrin expression. While lactoferrin was not expressed in adherent monolayer cultures under standard subconfluent conditions on plastic, lactoferrin mRNA and protein steadily accumulated when the cells aggregated to form spheroids on a reconstituted basement membrane gel. However, unlike other milk proteins such as beta-casein, lactoferrin expression was also induced at high cell density in the absence of exogenously added basement membrane or prolactin. These results led us to examine whether changes in cell growth, cell-cell interactions and/or cell shape were responsible for regulation of lactoferrin gene expression. Rounded, non-proliferating cells in suspension in serum-free medium expressed lactoferrin even as single cells. Conversely, lactoferrin expression could be inhibited in non-proliferative cells in serum-free medium by maintaining them in contact with an air-dried extracellular matrix which caused the cells to retain flat, spread morphologies. These findings indicated that cessation of cell growth was not sufficient, that cell-cell interactions were not required, and that cell culture conditions which minimize cell spreading may be important in maintaining lactoferrin expression. Additional data supporting this latter concept were generated by treating spread cells with cytochalasin D. The resulting disruption of microfilament assembly induced both cell rounding and lactoferrin expression. Shape-dependent regulation of lactoferrin mRNA was both transcriptional and post-transcriptional. Surprisingly, treatment of rounded cells with a transcription inhibitor, actinomycin D, produced a stabilization of lactoferrin mRNA, suggesting that transcription of an unstable factor is required for degradation of lactoferrin mRNA. Importantly, lactoferrin mRNA expression was regulated similarly in early passage normal human mammary epithelial cells. In vivo, the changing extracellular matrix components of the mammary gland during different stages of normal and abnormal growth and differentiation may provide different physical constraints on the configurations of cell surface molecules. These physical constraints may be communicated to the cell interior through mechanical changes in the cytoskeleton. Unlike beta-casein whose expression is upregulated by specific integrin-mediated signals, lactoferrin may be representative of a class of proteins synthesized in the mammary gland using basal transcriptional and translational machinery. The suppression of lactoferrin expression that is observed in monolayer culture and in malignant tissues may reflect inappropriate cell shapes and cytoskeletal structures that are manifested under these conditions.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/ultraestructura , Células Epiteliales/metabolismo , Lactoferrina/biosíntesis , Transducción de Señal , Animales , Línea Celular , Tamaño de la Célula , Citoesqueleto/metabolismo , Células Epiteliales/ultraestructura , Regulación de la Expresión Génica , Humanos
16.
J Biol Chem ; 270(12): 6872-80, 1995 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-7896835

RESUMEN

To gain insight into the signal transduction pathways utilized by the Wnt-1-responsive mammary epithelial cell line C57MG, we screened for non-src family member tyrosine kinases expressed in these cells using a polymerase chain reaction-based technique. We identified five cDNA clones encoding receptor tyrosine kinases for which the ligand is known (fibroblast growth factor receptor, platelet-derived growth factor receptor, epithelial growth factor receptor, insulin receptor, and insulin-like growth factor receptor), two putative receptor tyrosine kinases for which the ligand remains to be identified (the products of ryk and the mouse klg homolog), and a novel tyrosine kinase. We cloned cDNAs encoding both the murine and human homologs of this kinase, the sequences of which were subsequently published under the names sky (Ohashi, K., Mizuno, K., Kuma, K., Miyata, T., and Nakamura, T. (1994) Oncogene 9, 699-705) and rse (Mark, M. R., Scadden, D. T., Wang, Z., Gu, Q., Goddard, A., and Godowski, P. J. (1994) J. Biol. Chem. 269, 10720-10728). Mouse sky RNA levels are abundant in mammary tumors derived from transgenic mice that express wnt-1, fgf-3, or both oncogenes in their mammary glands. However, little or no expression of sky is detected in mammary glands from virgin animals or in preneoplastic mammary glands from wnt-1 transgenic mice. Moreover, we find that the human homolog of sky is expressed at elevated levels when normal human mammary epithelial cells are rendered tumorigenic by the introduction of two viral oncogenes. Transient transfection of the human SKY cDNA into the quail fibrosarcoma cell line QT6 reveals that SKY is an active tyrosine kinase that augments the level of cellular phosphotyrosine. Introduction of murine Sky into RatB1a fibroblasts by retrovirus-mediated gene transfer results in morphological transformation, growth in soft agar, and the formation of tumors in nude mice. These data raise the possibility that the Sky tyrosine kinase is involved in the development and/or progression of mammary tumors.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Experimentales/metabolismo , ARN Mensajero/análisis , Proteínas Tirosina Quinasas Receptoras/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Transformación Celular Neoplásica , ADN Complementario/aislamiento & purificación , Glicosilación , Humanos , Glándulas Mamarias Animales/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Tirosina Quinasas Receptoras/análisis , Células Tumorales Cultivadas
17.
Eur J Biochem ; 226(2): 725-30, 1994 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-7528142

RESUMEN

A calmodulin-like protein, which is identical in size and 85% identical to vertebrate calmodulin, was recently identified by 'subtractive hybridization' comparison of transcripts expressed in normal versus transformed human mammary epithelial cells. Unlike the ubiquitous distribution of calmodulin, calmodulin-like protein expression is restricted to certain epithelial cells, and appears to be modulated during differentiation. In addition, calmodulin-like protein levels are often significantly reduced in malignant tumor cells as compared to corresponding normal epithelial cells. The current studies compare calmodulin-like protein functions with those of calmodulin. We find that calmodulin-like protein activation of multifunctional Ca2+/calmodulin-dependent protein kinase II (calmodulin kinase II) is equivalent to activation by calmodulin, but that four other calmodulin-dependent enzymes, cGMP phosphodiesterase, calcineurin, nitric-oxide synthase, and myosin-light-chain kinase, display much weaker activation by calmodulin-like protein than by calmodulin. In the case of myosin-light-chain kinase, calmodulin-like protein competitively inhibits calmodulin activation of the enzyme with a Ki value of 170 nM. Thus, calmodulin-like protein may have evolved to function as a specific agonist of certain calmodulin-dependent enzymes, and/or as a specific competitive antagonist of other calmodulin-dependent enzymes.


Asunto(s)
Proteínas de Unión al Calcio/farmacología , Calmodulina/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos , 3',5'-GMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Aminoácido Oxidorreductasas/antagonistas & inhibidores , Aminoácido Oxidorreductasas/metabolismo , Secuencia de Aminoácidos , Unión Competitiva , Mama , Calcineurina , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Calmodulina/química , Calmodulina/metabolismo , Proteínas de Unión a Calmodulina/antagonistas & inhibidores , Proteínas de Unión a Calmodulina/metabolismo , Células Cultivadas , Cromatografía de Afinidad , Epitelio/química , Femenino , Humanos , Datos de Secuencia Molecular , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Óxido Nítrico Sintasa , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo
19.
FEBS Lett ; 334(3): 301-8, 1993 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-8243637

RESUMEN

We tested the hypothesis that early alterations in calcium influx induced by an imposed 60 Hz magnetic field are propagated down the signal transduction (ST) cascade to alter c-MYC mRNa induction. To test this we measured both ST parameters in the same cells following 60 Hz magnetic field exposures in a specialized annular ring device (220 G (22 mT), 1.7 mV/cm maximal E(induced), 37 degrees C, 60 min). Ca2+ influx is a very early ST marker that precedes the specific induction of mRNA transcripts for the proto-oncogene c-MYC, an immediate early response gene. In three experiments influx of 45Ca2+ in the absence of mitogen was similar to that in cells treated with suboptimal levels of Con-A (1 micrograms/ml). However, calcium influx was elevated 1.5-fold when lymphocytes were exposed to Con-A plus magnetic fields; this co-stimulatory effect is consistent with previous reports from our laboratory [FEBS Lett. 301 (1992) 53-59; FEBS Lett. 271 (1990) 157-160; Ann. N.Y. Acad. Sci. 649 (1992) 74-95]. The level of c-MYC mRNA transcript copies in non-activated cells and in suboptimally-activated cells was also similar, which is consistent with the above calcium influx findings. Significantly, lymphocytes exposed to the combination of magnetic fields plus suboptimal Con-A responded with an approximate 3.0-fold increase in band intensity of c-MYC mRNA transcripts. Importantly, transcripts for the housekeeping gene GAPDH were not influenced by mitogen or magnetic fields. We also observed that lymphocytes that failed to exhibit increased calcium influx in response to magnetic fields plus Con-A, also failed to exhibit an increase in total copies of c-MYC mRNA. Thus, calcium influx and c-MYC mRNA expression, which are sequentially linked via the signal transduction cascade in contrast to GAPDH, were both increased by magnetic fields. These findings support the above ST hypothesis and provide experimental evidence for a general biological framework for understanding magnetic field interactions with the cell through signal transduction. In addition, these findings indicate that magnetic fields can act as a co-stimulus at suboptimal levels of mitogen; pronounced physiological changes in lymphocytes such as calcium influx and c-MYC mRNA induction were not triggered by a weak mitogenic signal unless accompanied by a magnetic field. Magnetic fields, thus, have the ability to potentiate or amplify cell signaling.


Asunto(s)
Calcio/metabolismo , Magnetismo , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Transducción de Señal , Animales , Northern Blotting , Inducción Enzimática , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Timo/metabolismo
20.
Exp Cell Res ; 208(1): 175-88, 1993 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7689475

RESUMEN

We demonstrate that blockage of EGF receptor signal transduction is sufficient by itself to cause a rapid, efficient, and reversible G0-like growth arrest of normal human mammary epithelial cells (HMEC) of finite lifespan as well as two immortally transformed cell lines derived from normal HMEC following in vitro transformation with benzo[a]pyrene. For normal HMEC, the significant level of endogenous production of TGF alpha requires utilization of blocking antibodies to the EGF receptor to achieve cessation of growth in mass culture, whereas removal of EGF is sufficient to arrest the immortal cell lines. In the growth-arrested cells, protein synthesis remains depressed; reexposure to EGF leads to a rapid increase in protein synthesis. Inhibition of DNA synthesis is not detectable until approximately 12 h after removal of EGF/TGF alpha and is pronounced by 24 h. Reexposure to EGF produces high levels of synthesis of the early response genes, c-myc, c-fos, c-jun, and MGSA, within 1 h. DNA synthesis increases only after 10 h, with a sharp peak after 15-20 h. Reexposure of the growth-arrested normal HMEC for 1 h with EGF allows a majority of the cells capable of cycling to subsequently enter the S phase. Little is currently known about cell cycle control in normal human epithelial cells. The efficient and gentle method of achieving reversible G0 growth arrest reported here may facilitate studies on the cell cycle of this cell type. Additionally, results from normal HMEC can be compared with those from syngeneic immortalized cell populations to determine possible cell cycle parameters altered as a result of immortal transformation.


Asunto(s)
Mama/citología , Ciclo Celular , Quimiocinas CXC , Receptores ErbB/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intercelular , Anticuerpos Monoclonales , Calmodulina/genética , Ciclo Celular/efectos de los fármacos , Quimiocina CXCL1 , Factores Quimiotácticos/genética , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales , Expresión Génica/efectos de los fármacos , Genes fos , Genes jun , Genes myc , Sustancias de Crecimiento/genética , Histonas/genética , Humanos , Técnicas In Vitro , Queratinas/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA