Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
Oncogene ; 35(47): 6053-6064, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27157621

RESUMEN

Amplification and overexpression of erbB2/neu proto-oncogene is observed in 20-30% human breast cancer and is inversely correlated with the survival of the patient. Despite this, somatic activating mutations within erbB2 in human breast cancers are rare. However, we have previously reported that a splice isoform of erbB2, containing an in-frame deletion of exon 16 (herein referred to as ErbB2ΔEx16), results in oncogenic activation of erbB2 because of constitutive dimerization of the ErbB2 receptor. Here, we demonstrate that the ErbB2ΔEx16 is a major oncogenic driver in breast cancer that constitutively signals from the cell surface. We further show that inducible expression of the ErbB2ΔEx16 variant in mammary gland of transgenic mice results in the rapid development of metastatic multifocal mammary tumors. Genetic and biochemical characterization of the ErbB2ΔEx16-derived mammary tumors exhibit several unique features that distinguish this model from the conventional ErbB2 ones expressing the erbB2 proto-oncogene in mammary epithelium. Unlike the wild-type ErbB2-derived tumors that express luminal keratins, ErbB2ΔEx16-derived tumors exhibit high degree of intratumoral heterogeneity co-expressing both basal and luminal keratins. Consistent with these distinct pathological features, the ErbB2ΔEx16 tumors exhibit distinct signaling and gene expression profiles that correlate with activation of number of key transcription factors implicated in breast cancer metastasis and cancer stem cell renewal.


Asunto(s)
Empalme Alternativo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/genética , Receptor ErbB-2/genética , Microambiente Tumoral/genética , Animales , Línea Celular Tumoral , Análisis por Conglomerados , Modelos Animales de Enfermedad , Exones , Matriz Extracelular/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neoplasias Pulmonares/secundario , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia , Fenotipo , Proto-Oncogenes Mas , Eliminación de Secuencia , Factores de Transcripción/metabolismo
2.
BMC Cancer ; 15: 562, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26228788

RESUMEN

BACKGROUND: The mammary glands of pigs share many functional and morphological similarities with the breasts of humans, raising the potential of their utility for research into the mechanisms underlying normal mammary function and breast carcinogenesis. Here we sought to establish a model for the efficient manipulation and transformation of porcine mammary epithelial cells (pMEC) in vitro and tumor growth in vivo. METHODS: We utilized a vector encoding the red florescent protein tdTomato to transduce populations of pMEC from Yorkshire -Hampshire crossbred female pigs in vitro and in vivo. Populations of primary pMEC were then separated by FACS using markers to distinguish epithelial cells (CD140a-) from stromal cells (CD140a+), with or without further enrichment for basal and luminal progenitor cells (CD49f+). These separated pMEC populations were transduced by lentivirus encoding murine polyomavirus T antigens (Tag) and tdTomato and engrafted to orthotopic or ectopic sites in immunodeficient NOD.Cg-Prkdc (scid) Il2rg (tm1Wjl) /SzJ (NSG) mice. RESULTS: We demonstrated that lentivirus effectively transduces pMEC in vitro and in vivo. We further established that lentivirus can be used for oncogenic-transformation of pMEC ex vivo for generating mammary tumors in vivo. Oncogenic transformation was confirmed in vitro by anchorage-independent growth, increased cell proliferation, and expression of CDKN2A, cyclin A2 and p53 alongside decreased phosphorylation of Rb. Moreover, Tag-transformed CD140a- and CD140a-CD49f + pMECs developed site-specific tumors of differing histopathologies in vivo. CONCLUSIONS: Herein we establish a model for the transduction and oncogenic transformation of pMEC. This is the first report describing a porcine model of mammary epithelial cell tumorigenesis that can be applied to the study of human breast cancers.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Transformación Celular Viral/genética , Lentivirus/genética , Glándulas Mamarias Animales/trasplante , Neoplasias Mamarias Experimentales/patología , Poliomavirus/inmunología , Animales , Proliferación Celular , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Técnicas In Vitro , Lentivirus/fisiología , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/virología , Neoplasias Mamarias Experimentales/etiología , Poliomavirus/genética , Porcinos
3.
Oncogene ; 34(29): 3815-25, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25263440

RESUMEN

The biological outcome of TMPRSS2:ERG chromosomal translocations in prostate cancer (PC) remains poorly understood. To address this, we compared the transcriptional effects of TMPRSS2:ERG expression in a transgenic mouse model with those of ERG knockdown in a TMPRSS2:ERG-positive PC cell line. This reveals that ERG represses the expression of a previously unreported set of androgen receptor (AR)-independent neuronal genes that are indicative of neuroendocrine (NE) cell differentiation-in addition to previously reported AR-regulated luminal genes. Cell sorting and proliferation assays performed after sustained ERG knockdown indicate that ERG drives proliferation and blocks the differentiation of prostate cells to both NE and luminal cell types. Inhibition of ERG expression in TMPRSS2:ERG-positive PC cells through blockade of AR signaling is tracked with increased NE gene expression. We also provide evidence that these NE cells are resistant to pharmacological AR inhibition and can revert to the phenotype of parental cells upon restoration of AR/ERG signaling. Our findings highlight an ERG-regulated mechanism capable of repopulating the parent tumor through the transient generation of an anti-androgen therapy-resistant cell population, suggesting that ERG may have a direct role in preventing resistance to anti-androgen therapy.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Neoplasias de la Próstata/genética , Serina Endopeptidasas/genética , Transactivadores/genética , Animales , Antibióticos Antineoplásicos/farmacología , Benzamidas , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Doxorrubicina/farmacología , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Ratones Transgénicos , Microscopía Confocal , Células Neuroendocrinas/metabolismo , Nitrilos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Interferencia de ARN , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina Endopeptidasas/metabolismo , Transactivadores/metabolismo , Regulador Transcripcional ERG , Translocación Genética
4.
Oncogene ; 33(50): 5729-39, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-24317513

RESUMEN

Pregnancy-associated breast cancers (PABCs) are tumors diagnosed during pregnancy or up to 5 years following parturition, and are usually high-grade, connective tissue-rich, and estrogen receptor (ER)/progesterone receptor-negative. Little is known about the cellular origin of PABCs or the mechanisms by which PABCs are initiated. Using the RCAS retrovirus to deliver the ErbB2 oncogene into the mammary epithelium of our previously reported MMTV-tva transgenic mice, we detected high-grade, poorly differentiated, stroma-rich and ER-negative tumors during pregnancy and lactation. These high-grade and stroma-rich tumors were less frequent in involuted mice or in age-matched nulliparous mice. More importantly, by generating a WAP-tva transgenic line for expression of ErbB2 selectively in WAP(+) mammary alveolar cells, we found that tumors had similar morphological phenotypes (high grade, poorly differentiated, stroma-rich and ER-negative), irrespective of the time since pregnancy and even in the absence of pregnancy. These data suggest that PABCs arise preferentially from an alveolar cell population that expands during pregnancy and lactation. This somatic mouse model may also be useful for preclinical testing of new prophylactic and therapeutic strategies against PABC.


Asunto(s)
Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/patología , Células Madre Neoplásicas/patología , Complicaciones Neoplásicas del Embarazo/patología , Animales , Transformación Celular Viral , Femenino , Humanos , Ratones , Ratones Transgénicos , Clasificación del Tumor , Células Madre Neoplásicas/fisiología , Fenotipo , Embarazo
5.
Oncogenesis ; 2: e74, 2013 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-24080956

RESUMEN

The phosphoinositide 3-kinase (PI3K) signaling cascade is a key mediator of cellular growth, survival and metabolism and is frequently subverted in human cancer. The gene encoding for the alpha catalytic subunit of PI3K (PIK3CA) is mutated and/or amplified in ∼30% of breast cancers. Mutations in either the kinase domain (H1047R) or the helical domain (E545K) are most common and result in a constitutively active enzyme with oncogenic capacity. PIK3CA(H1047R) was previously demonstrated to induce tumors in transgenic mouse models; however, it was not known whether overexpression of PIK3CA(E545K) is sufficient to induce mammary tumors and whether tumor initiation by these two types of mutants differs. Here, we demonstrate that expression of PIK3CA(E545K) in the mouse mammary gland induces heterogenous mammary carcinomas but with a longer latency than PIK3CA(H1047R)-expressing mice. Our results suggest that the helical domain mutant PIK3CA(E545K) is a less potent inducer of mammary tumors due to less efficient activation of downstream Akt signaling.

6.
Oncogene ; 31(20): 2545-54, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21996730

RESUMEN

A hallmark of human cancer is heterogeneity, reflecting the complex series of changes resulting in the activation of oncogenes coupled with inactivation of tumor suppressor genes. Breast cancer is no exception and indeed, many studies have revealed considerable complexity and heterogeneity in the population of primary breast tumors and substantial changes in a recurrent breast tumor that has acquired metastatic properties and drug resistance. We have made use of a Myc-inducible transgenic mouse model of breast cancer in which elimination of Myc activity following tumor development initially leads to a regression of a subset of tumors generally followed by de novo Myc-independent growth. We have observed that tumors that grow independent of Myc expression have gene profiles that are distinct from the primary tumors with characteristics indicative of an epithelial-mesenchymal transition (EMT) phenotype. Phenotypic analyses of Myc-independent tumors confirm the acquisition of an EMT phenotype suggested to be associated with invasive and migratory properties in human cancer cells. Further genomic analyses reveal mouse mammary tumors growing independent of myc have a higher probability of exhibiting a gene signature similar to that observed for human 'tumor-initiating' cells. Collectively, the data reveal genetic alterations that underlie tumor progression and an escape from Myc-dependent growth in a transgenic mouse model that can provide insights to what occurs in human cancers as they acquire drug resistance and metastatic properties.


Asunto(s)
Neoplasias de la Mama , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/genética , Neoplasias Mamarias Experimentales , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes ras , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Células Madre Neoplásicas/patología , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador beta/genética
7.
Vet Pathol ; 49(1): 116-29, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22173978

RESUMEN

Neoplasia in both animals and humans results in part from lasting activation of tumor-promoting genes ("oncogenes") or diminished function of genes responsible for preventing neoplastic induction ("tumor suppressor genes"). The concept of "genetic addiction" has emerged to indicate that neoplastic cells cannot maintain a malignant phenotype without sustained genotypic abnormalities related to aberrant activity of oncogene(s) and/or inactivity of tumor suppressor gene(s). Interestingly, some genetic abnormalities reliably produce distinct morphologic patterns that can be used as structural signatures indicating the presence of a specific molecular alteration. Examples of such consistent genetic/microanatomic pairings have been identified for mutated oncogenes, such as rising mucin-producing capacity with RAS overexpression, and mutated tumor suppressor genes-including PTEN eliciting cell hypertrophy, RB1 dictating neuroendocrine differentiation, and TRP53 encouraging sarcomatous transformation. Familiarity with the concept of genetic addiction, as well as the ability to recognize such regular genomic-phenotypic relationships, are of paramount importance for comparative pathologists who are engaged in phenotyping genetically engineered mice to help unravel genomic intricacies in both health and disease.


Asunto(s)
Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica/genética , Genotipo , Neoplasias/genética , Fenotipo , Animales , Modelos Animales de Enfermedad , Genes Supresores de Tumor , Ingeniería Genética , Humanos , Ratones , Ratones Transgénicos , Mutación , Neoplasias/patología , Oncogenes
8.
Oncogene ; 30(3): 301-12, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20818417

RESUMEN

C-Src is infrequently mutated in human cancers but it mediates oncogenic signals of many activated growth factor receptors and thus remains a key target for cancer therapy. However, the broad function of Src in many cell types and processes requires evaluation of Src-targeted therapeutics within a normal developmental and immune-competent environment. In an effort to understand the appropriate clinical use of Src inhibitors, we tested an Src inhibitor, SKI-606 (bosutinib), in the MMTV-PyVmT transgenic mouse model of breast cancer. Tumor formation in this model is dependent on the presence of Src, but the necessity of Src kinase activity for tumor formation has not been determined. Furthermore, Src inhibitors have not been examined in an autochthonous tumor model that permits assessment of effects on different stages of tumor progression. Here we show that oral administration of SKI-606 inhibited the phosphorylation of Src in mammary tumors and caused a rapid decrease in the Ezh2 Polycomb group histone H3K27 methyltransferase and an increase in epithelial organization. SKI-606 prevented the appearance of palpable tumors in over 50% of the animals and stopped tumor growth in older animals with pre-existing tumors. These antitumor effects were accompanied by decreased cellular proliferation, altered tumor blood vessel organization and dramatically increased differentiation to lactational and epidermal cell fates. SKI-606 controls the development of mammary tumors by inducing differentiation.


Asunto(s)
Compuestos de Anilina/farmacología , Diferenciación Celular/efectos de los fármacos , Neoplasias Mamarias Experimentales/patología , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Animales , Femenino , Perfilación de la Expresión Génica , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Transgénicos
9.
Vet Pathol ; 46(4): 736-45, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19276050

RESUMEN

Prolactin-secreting pituitary adenomas are common spontaneous lesions in aging FVB females. Prolactin-secreting pituitary proliferations play a significant role in mouse mammary tumorigenesis generally producing adenosquamous carcinomas. Since genetically engineered FVB mice are frequently used to study mammary tumor biology, we have examined a cohort of 64 aging wild-type FVB/N females to establish the prevalence and the nature of spontaneous mammary and pituitary tumors. Tissues from mammary and pituitary glands were studied by histopathology and immunohistochemistry. Of the 64 examined mice, 20 had pituitary tumors and 20 had mammary tumors. Mammary and pituitary tumors were associated in 17 mice. All pituitary tumors were prolactin-positive by immunohistochemistry and classified as prolactinomas. Fourteen mammary tumors, including 12 cases with and 2 without concurrent prolactinomas, were adenocarcinomas with different combinations of epithelial growth patterns. Five mice with prolactinomas had mammary tumors characterized by the epithelial-mesenchymal transition (EMT) phenotype. Estrogen receptor alpha (ERalpha)-positivity was observed for 14 of the 18 mammary tumors tested, including both adenocarcinomas with nuclear immunoreactivity and EMT-phenotype tumors with both nuclear and cytoplasmic immunoreactivity. No immunoreactivity for the progesterone receptor was observed. This study confirms that spontaneous prolactinomas and mammary tumors are both common and significantly associated lesions in FVB mice. Parity and age represented risk factors for the development of these tumors. Compared with previous reports, prolactinoma-associated mammary tumors displayed a broader morphologic spectrum, including cases with the EMT phenotype. The elevated number of prolactinoma-associated and ERalpha-positive mammary tumors opens intriguing possibilities concerning the role of ERalpha cytoplasmic localization during EMT tumorigenesis.


Asunto(s)
Neoplasias Mamarias Animales/epidemiología , Neoplasias Mamarias Animales/patología , Ratones Transgénicos , Fenotipo , Neoplasias Hipofisarias/epidemiología , Prolactinoma/epidemiología , Factores de Edad , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Neoplasias Mamarias Animales/etiología , Ratones , Neoplasias Hipofisarias/complicaciones , Prevalencia , Prolactinoma/complicaciones , Factores de Riesgo
10.
Artículo en Inglés | MEDLINE | ID: mdl-16869764

RESUMEN

Because the pRb pathway is disrupted in most solid human cancers, we have generated genetically engineered mouse cancer models by inactivating pRb function in several cell types, including astrocytes and mammary, prostate, ovarian, and brain choroid plexus epithelia. In every case, proliferation and apoptosis are acutely induced, predisposing to malignancy. Cell type dictates the pathways involved in tumor progression. In the astrocytoma model, we developed strategies to induce events in the adult brain, either throughout the tissue or focally. Both K-Ras activation and Pten inactivation play significant roles in progression. In the prostate model, adenocarcinoma progression depends on Pten inactivation. However, nonautonomous induction of p53 in the mesenchyme leads to evolution of both compartments, with p53 loss occurring in the mesenchyme. Thus, studies in these models continue to identify key tumorigenesis mechanisms. Furthermore, we are hopeful that the models will provide useful preclinical systems for diagnostic and therapeutic development.


Asunto(s)
Neoplasias Experimentales/genética , Animales , Astrocitoma/etiología , Astrocitoma/genética , Astrocitoma/patología , Femenino , Genes de Retinoblastoma , Genes ras , Ingeniería Genética , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteína de Retinoblastoma/antagonistas & inhibidores
11.
Curr Drug Targets ; 4(3): 263-79, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12643476

RESUMEN

Animal models of prostate cancer have been limited in number and in relevance to the human disease. With the advancement of transgenic and knockout technologies, combined with tissue specific promoters and tissue-specific gene ablation, a new generation of mouse models has emerged. This review will discuss various animal models and their inherent strengths and weaknesses. A primary emphasis is placed on mouse models that have been designed on the basis of genetic alterations that are frequently found in human prostate cancer. These models display slow, temporal development of increasingly severe histopathologic lesions, which are remarkably restricted to the prostate gland, a property similar to the ageing related progression of this disease in humans. The preneoplastic lesions, akin to what is considered as prostatic intraepithelial neoplasia, are consistent major phenotypes in the models, and, therefore. are discussed for histopathologic criteria that may distinguish their progressions or grades. Finally, considering that prostate cancer is a complex multifocal disease, which is likely to require multiple genetic/epigenetic alterations, many of these models have already been intercrossed to derive mice with compound genetic alterations. It is predicted that these and subsequent compound mutant mice should represent "natural" animal models for investigating the mechanism of development of human prostate diseases, as well as, for preclinical models for testing therapeutics.


Asunto(s)
Modelos Animales de Enfermedad , Neoplasias de la Próstata/genética , Animales , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neoplasias de la Próstata/patología , Ratas
12.
Cancer Res ; 61(24): 8811-9, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11751403

RESUMEN

A coordinated growth arrest during mammary involution completes the dramatic changes in mammary cell proliferation seen during pregnancy and lactation. Signals regulating this arrest are poorly understood, despite their potential relevance to oncogenesis. Here we report that the arrest involves a unique pulse of p16(INK4A) expression in vivo, which accompanies decreased cyclin D1 expression and a shift to an active repressor E2F4 complex. We used INK4A/ARF-/- mice as well as cyclin D1 and p16(INK4A) transgenic strains to examine the physiological significance of these patterns. p16(INK4A) directly regulated the in vivo transition from E2F3 to E2F4 as the major E2F DNA binding activity, and its contribution to growth arrest was independent of cyclin D1. Transgenic cyclin D1 expression prevented normal terminal differentiation by ablating the p16(INK4A) pulse, abolishing the shift from E2F3 to E2F4, derepressing E2F target genes, and expanding a stem cell population. The effects of cyclin D1 were reversed by restoring p16(INK4A) but were not seen in INK4A/ARF-/- mice. Our results indicate that cyclin D1 may contribute to tumorigenesis by altering cell differentiation and demonstrate a significant function for p16(INK4A) in development in vivo. These regulatory mechanisms used during mammary involution offer a potential explanation for the protective effect of pregnancy against breast cancer.


Asunto(s)
Ciclina D1/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Glándulas Mamarias Animales/crecimiento & desarrollo , Células 3T3 , Animales , Diferenciación Celular/genética , División Celular/fisiología , Ciclina D1/biosíntesis , Ciclina D1/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Vectores Genéticos/genética , Humanos , Lactancia/metabolismo , Lactancia/fisiología , Glándulas Mamarias Animales/fisiología , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Transgénicos , Embarazo , Células Tumorales Cultivadas
13.
Oncogene ; 20(48): 7064-72, 2001 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-11704830

RESUMEN

The integrin linked kinase (ILK) is a cytoplasmic effector of integrin receptors, involved in the regulation of integrin binding properties as well as the activation of cell survival and proliferative pathways, including those involving MAP kinase, PKB/Akt and GSK-3beta. Overexpression of ILK in cultured intestinal and mammary epithelial cells has been previously shown to induce changes characteristic of oncogenic transformation, including anchorage-independent growth, invasiveness, suppression of anoikis and tumorigenicity in nude mice. In order to determine if ILK overexpression can result in the formation of mammary tumors in vivo, we generated transgenic mice expressing ILK in the mammary epithelium, under the transcriptional control of the mouse mammary tumor virus (MMTV) long terminal repeat (LTR). By the age of 6 months, female MMTV/ILK mice developed a hyperplastic mammary phenotype, which was accompanied by the constitutive phosphorylation of PKB/Akt, GSK-3beta and MAP kinase. Focal mammary tumors subsequently appeared in 34% of the animals at an average age of 18 months. Given the focal nature and long latency of the tumors, however, additional genetic events are likely required for tumor induction in the MMTV/ILK mice. These results provide the first direct demonstration of a potential oncogenic role for ILK, which is upregulated in human tumors and tumor cell lines.


Asunto(s)
Mama/patología , Neoplasias Mamarias Experimentales/enzimología , Proteínas de Neoplasias/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Adenocarcinoma Papilar/enzimología , Adenocarcinoma Papilar/genética , Animales , Mama/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Transformación Celular Neoplásica/genética , Inducción Enzimática , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3 , Humanos , Hiperplasia , Integrinas/metabolismo , Sistema de Señalización de MAP Quinasas , Neoplasias Mamarias Experimentales/genética , Virus del Tumor Mamario del Ratón/genética , Metaplasia , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Especificidad de Órganos , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Recombinantes de Fusión/fisiología , Secuencias Repetidas Terminales/genética , Transgenes
14.
Transgenic Res ; 10(5): 471-8, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11708657

RESUMEN

Overexpression of the cyclin D1 oncogene and inactivation of the p53 tumor suppressor have both been implicated in substantial proportions of sporadic human breast cancers. Transgenic mice with cyclin D1 overexpression targeted to mammary tissue by the MMTV enhancer-promoter have been shown to develop mammary cancers. To investigate the relationship between pathways driven by cyclin D1 overexpression and p53 loss during the development of breast cancers, we crossed MMTV-cyclin D1 mice with p53 heterozygous null (p53+/-) mice. In such crossed mice, cyclin D1-driven mammary neoplasia would need to be substantially accelerated by p53 loss in order for mammary tumors to develop prior to the expected onset of non-mammary tumors characteristic of the p53-deficient background alone. Instead, in mice heterozygous or homozygous for p53 deficiency and simultaneously carrying the MMTV-cyclin D1 transgene, only tumors typically found in p53-deficient mice developed and mammary tumors were not observed. Interestingly, MMTV-cyclin D1/p53+/- mice appeared to develop these non-mammary tumors more rapidly than p53+/- mice, and a majority of the sampled non-mammary tumors from MMTV-cyclin D1/p53+/- mice showed 'ectopic' expression of the MMTV-driven cyclin D1 transgene. Within the constraints of possible genetic background effects and limited sensitivity due to the early emergence of non-mammary tumors, these observations provide no evidence that inactivation of p53 confers a major additional selective advantage to mammary cells overexpressing cyclin D1 in this animal model of human breast cancer. Interestingly, the results do raise the possibility that p53 inactivation might complement or cooperate with cyclin D1 deregulation during the development of some types of non-mammary tumors.


Asunto(s)
Ciclina D1/genética , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/genética , Virus del Tumor Mamario del Ratón/genética , Proteína p53 Supresora de Tumor/genética , Animales , Femenino , Glándulas Mamarias Animales/fisiología , Neoplasias Mamarias Animales/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Neoplasias/genética , Neoplasias/patología
15.
Cancer Res ; 61(22): 8298-305, 2001 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11719463

RESUMEN

Mice transgenic for the Polyomavirus middle T (PyV-mT) gene have been widely used to study mammary tumorigenesis and metastasis. Although numerous molecular insights were gained from the analysis of these transgenic malignant tumors, the early events leading to malignant transformation have not been systematically investigated nor has the biological potential of hyperplastic lesions been documented. This paper presents the first comprehensive histopathological characterization of transgenic PyV-mT hyperplasias together with classical transplantation experiments designed to test the growth potential of these lesions. Moreover, stable hyperplastic outgrowth lines were established as a tool to study premalignant PyV-mT-induced hyperplasias in detail. Each line has a different tumor latency, indicating that PyV-mT-induced hyperplasias, like early proliferative lesions seen in the human breast, are heterogeneous with respect to their malignant potential. Our results settle a controversy; they establish that PyV-mT gene expression alone is insufficient to induce tumors and that additional events are required for tumorigenesis and metastasis. These results support the use of PyV-mT transgenic mice as a model for investigating the multistep progression of malignant mammary tumorigenesis and metastasis.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Animales , Antígenos Transformadores de Poliomavirus/biosíntesis , División Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/metabolismo , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Lesiones Precancerosas/irrigación sanguínea , Lesiones Precancerosas/metabolismo , Receptores de Estrógenos/biosíntesis , Receptores de Progesterona/biosíntesis , Proteína p53 Supresora de Tumor/biosíntesis
16.
Mol Cell Biol ; 21(24): 8547-64, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11713289

RESUMEN

E2F activity is critical for the control of the G(1) to S phase transition. We show that the combined loss of E2F1 and E2F2 results in profound effects on hematopoietic cell proliferation and differentiation, as well as increased tumorigenesis and decreased lymphocyte tolerance. The loss of E2F1 and E2F2 impedes B-cell differentiation, and hematopoietic progenitor cells in the bone marrow of mice lacking E2F1 and E2F2 exhibit increased cell cycling. Importantly, we show that E2F1 and E2F2 double-knockout T cells exhibit more rapid entry into S phase following antigenic stimulation. Furthermore, T cells lacking E2F1 and E2F2 proliferate much more extensively in response to subthreshold antigenic stimulation. Consistent with these observations, E2F1/E2F2 mutant mice are highly predisposed to the development of tumors, and some mice exhibit signs of autoimmunity.


Asunto(s)
Antígenos/metabolismo , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Neoplasias/metabolismo , Linfocitos T/citología , Linfocitos T/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Factores de Edad , Animales , Western Blotting , Bromodesoxiuridina/metabolismo , Diferenciación Celular , División Celular , Células Cultivadas , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Factor de Transcripción E2F2 , Femenino , Citometría de Flujo , Genotipo , Ganglios Linfáticos/citología , Linfocitos/metabolismo , Masculino , Ratones , Ratones Noqueados , Ribonucleasas/metabolismo , Bazo/citología , Factores de Tiempo
18.
Oncogene ; 20(25): 3247-57, 2001 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-11423974

RESUMEN

Protein kinase CK2 is a ubiquitous and evolutionarily conserved serine/threonine kinase that is upregulated in many human cancers and can serve as an oncogene in lymphocytes. Recently, we have demonstrated that CK2 potentiates Wnt/beta-catenin signaling in mammary epithelial cells. To determine whether CK2 overexpression contributes to mammary tumorigenesis, we have performed comparative studies of human and rat breast cancer specimens and we have engineered transgenic mice with dysregulated expression of CK2alpha in the mammary gland. We find that CK2 is highly expressed in human breast tumor specimens and in carcinogen-induced rat mammary tumors. Overexpression of CK2alpha in the mammary gland of transgenic mice, under control of the MMTV-LTR, causes hyperplasia and dysplasia of the female mammary gland. Thirty per cent of the female MMTV-CK2alpha transgenic mice develop mammary adenocarcinomas at a median of 23 months of age, often associated with Wnt pathway activation, as evidenced by upregulation of beta-catenin protein. NF-kappaB activation and upregulation of c-Myc also occur frequently. Thus, in mice, rats, and humans, dysregulated expression of CK2 is associated with and is capable of contributing to mammary tumorigenesis. Targeted inhibition of CK2 could be useful in the treatment of breast cancer.


Asunto(s)
Adenocarcinoma/etiología , Neoplasias de la Mama/etiología , Transformación Celular Neoplásica , Neoplasias Mamarias Animales/etiología , Proteínas Serina-Treonina Quinasas/biosíntesis , Transactivadores , Proteínas de Pez Cebra , Edad de Inicio , Animales , Quinasa de la Caseína II , Proteínas del Citoesqueleto/metabolismo , Femenino , Enfermedad Fibroquística de la Mama , Humanos , Hiperplasia , Lactancia , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/inducido químicamente , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Proteínas Wnt , beta Catenina
19.
Cancer Res ; 61(8): 3480-5, 2001 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11309311

RESUMEN

Treatment of female C57BL/6J (B6) mice carrying the mutant Min allele of the adenomatous polyposis coli (Apc) gene with ethylnitrosourea (ENU) results in approximately 90% of mice developing an average of three mammary tumors within 65 days. As a first step in the identification of loci modifying susceptibility to ENU-induced mammary tumors and hyperplasias, we have tested ENU-treated Apc(Min)/+ (Min/+) mice on several hybrid backgrounds for susceptibility to mammary and intestinal tumors. C57BR/cdJxB6 (BRB6) Min/+ mice were more sensitive to development of mammary squamous cell carcinomas than B6 Min/+ mice. In contrast, Min/+ hybrids between B6 and FVB/NTac (FVB), 129X1/SvJ (129X1), and 129S6/SvEvTac (129S6) were all significantly more resistant to mammary carcinoma development. However, mice from these three crosses developed more focal mammary hyperplasias than did the B6 or BRB6 Min/+ mice. Susceptibility to intestinal tumors was independent of mammary tumor susceptibility in most hybrids. These results indicate that genetic background can affect independently the phenotypes conferred by the Min allele of APC:


Asunto(s)
Genes APC/genética , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Alelos , Animales , Cruzamientos Genéticos , Femenino , Predisposición Genética a la Enfermedad , Hiperplasia/genética , Neoplasias Intestinales/genética , Masculino , Ratones , Ratones Endogámicos C57BL
20.
Cancer Res ; 61(5): 1816-9, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11280729

RESUMEN

Werner syndrome is an autosomal recessive disorder characterized by genomic instability and by the premature onset of a number of age-related diseases, including malignancy. To assess a potential collaboration between p21 or p53 cell cycle regulators and Wrn proteins, Wrn mutant mice were created and mated with p21 or p53 null mice to generate double mutants. The p21 null/Wrn mutant mice did not show an acceleration of tumorigenesis during the first year of life, suggesting that the p53-dependent G1-S cell cycle checkpoint (which operates via p21) is not involved in Wrn-abetted tumor suppression. In contrast, the p53 null/Wrn mutant mice were particularly remarkable with respect to the rapidity with which they developed tumors. These mice were also distinguished by the variety of tumors they developed compared to those that developed in p53 null mice. Such data suggest a genetic interaction between p53 and Wrn in which loss of Wrn provokes a more variable p53 response unrelated to its role in the G1-S cell cycle checkpoint.


Asunto(s)
Ciclinas/fisiología , ADN Helicasas/genética , Mutación , Neoplasias Experimentales/genética , Proteína p53 Supresora de Tumor/fisiología , Síndrome de Werner/genética , Adenosina Trifosfatasas/genética , Animales , Cruzamientos Genéticos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Genes p53/genética , Hemangiosarcoma/enzimología , Hemangiosarcoma/genética , Ratones , Ratones Mutantes , Neoplasias Experimentales/enzimología , Estructura Terciaria de Proteína , RecQ Helicasas , Proteína p53 Supresora de Tumor/genética , Síndrome de Werner/complicaciones , Síndrome de Werner/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA