Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 80(17): 3631-3648, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32586983

RESUMEN

Pygopus 2 (Pygo2) is a coactivator of Wnt/ß-catenin signaling that can bind bi- or trimethylated lysine 4 of histone-3 (H3K4me2/3) and participate in chromatin reading and writing. It remains unknown whether the Pygo2-H3K4me2/3 association has a functional relevance in breast cancer progression in vivo. To investigate the functional relevance of histone-binding activity of Pygo2 in malignant progression of breast cancer, we generated a knock-in mouse model where binding of Pygo2 to H3K4me2/3 was rendered ineffective. Loss of Pygo2-histone interaction resulted in smaller, differentiated, and less metastatic tumors, due, in part, to decreased canonical Wnt/ß-catenin signaling. RNA- and ATAC-sequencing analyses of tumor-derived cell lines revealed downregulation of TGFß signaling and upregulation of differentiation pathways such as PDGFR signaling. Increased differentiation correlated with a luminal cell fate that could be reversed by inhibition of PDGFR activity. Mechanistically, the Pygo2-histone interaction potentiated Wnt/ß-catenin signaling, in part, by repressing the expression of Wnt signaling antagonists. Furthermore, Pygo2 and ß-catenin regulated the expression of miR-29 family members, which, in turn, repressed PDGFR expression to promote dedifferentiation of wild-type Pygo2 mammary epithelial tumor cells. Collectively, these results demonstrate that the histone binding function of Pygo2 is important for driving dedifferentiation and malignancy of breast tumors, and loss of this binding activates various differentiation pathways that attenuate primary tumor growth and metastasis formation. Interfering with the Pygo2-H3K4me2/3 interaction may therefore serve as an attractive therapeutic target for metastatic breast cancer. SIGNIFICANCE: Pygo2 represents a potential therapeutic target in metastatic breast cancer, as its histone-binding capability promotes ß-catenin-mediated Wnt signaling and transcriptional control in breast cancer cell dedifferentiation, EMT, and metastasis.


Asunto(s)
Desdiferenciación Celular/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Mamarias Experimentales/patología , Animales , Progresión de la Enfermedad , Femenino , Técnicas de Sustitución del Gen , Ratones , Ratones Endogámicos C57BL
2.
Oncogenesis ; 7(9): 73, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30237500

RESUMEN

Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase that regulates a plethora of downstream signaling pathways essential for cell migration, proliferation and death, processes that are exploited by cancer cells during malignant progression. These well-established tumorigenic activities, together with its high expression and activity in different cancer types, highlight FAK as an attractive target for cancer therapy. We have assessed and characterized the therapeutic potential and the biological effects of BI 853520, a novel small chemical inhibitor of FAK, in several preclinical mouse models of breast cancer. Treatment with BI 853520 elicits a significant reduction in primary tumor growth caused by an anti-proliferative activity by BI 853520. In contrast, BI 853520 exerts effects with varying degrees of robustness on the different stages of the metastatic cascade. Together, the data demonstrate that the repression of FAK activity by the specific FAK inhibitor BI 853520 offers a promising anti-proliferative approach for cancer therapy.

3.
Cell Rep ; 15(6): 1161-74, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27134168

RESUMEN

Despite the approval of several anti-angiogenic therapies, clinical results remain unsatisfactory, and transient benefits are followed by rapid tumor recurrence. Here, we demonstrate potent anti-angiogenic efficacy of the multi-kinase inhibitors nintedanib and sunitinib in a mouse model of breast cancer. However, after an initial regression, tumors resume growth in the absence of active tumor angiogenesis. Gene expression profiling of tumor cells reveals metabolic reprogramming toward anaerobic glycolysis. Indeed, combinatorial treatment with a glycolysis inhibitor (3PO) efficiently inhibits tumor growth. Moreover, tumors establish metabolic symbiosis, illustrated by the differential expression of MCT1 and MCT4, monocarboxylate transporters active in lactate exchange in glycolytic tumors. Accordingly, genetic ablation of MCT4 expression overcomes adaptive resistance against anti-angiogenic therapy. Hence, targeting metabolic symbiosis may be an attractive avenue to avoid resistance development to anti-angiogenic therapy in patients.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Resistencia a Antineoplásicos , Neoplasias Mamarias Animales/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Glucólisis/efectos de los fármacos , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/patología , Ratones , Modelos Biológicos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Neovascularización Patológica/patología
4.
Angiogenesis ; 19(3): 339-58, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27038485

RESUMEN

UNLABELLED: Viral VEGF-E (ovVEGF-E), a homolog of VEGF-A, was discovered in the genome of Orf virus. Together with VEGF-A, B, C, D, placental growth factor (PlGF) and snake venom VEGF (svVEGF), ovVEGF-E is a member of the VEGF family of potent angiogenesis factors with a bioactivity similar to VEGF-A: it induces proliferation, migration and sprouting of cultured vascular endothelial cells and proliferative lesions in the skin of sheep, goat and man that are characterized by massive capillary proliferation and dilation. These biological functions are mediated exclusively via its interaction with VEGF receptor-2 (VEGFR-2). Here, we have generated transgenic mice specifically expressing ovVEGF-E in ß-cells of the endocrine pancreas (Rip1VEGF-E; RVE). RVE mice show an increase in number and size of the islets of Langerhans and a distorted organization of insulin and glucagon-expressing cells. Islet endothelial cells of RVE mice hyper-proliferate and form increased numbers of functional blood vessels. In addition, the formation of disorganized lymphatic vessels and increased immune cell infiltration is observed. Upon crossing RVE single-transgenic mice with Rip1Tag2 (RT2) transgenic mice, a well-studied model of pancreatic ß-cell carcinogenesis, double-transgenic mice (RT2;RVE) display hyper-proliferation of endothelial cells resulting in the formation of hemangioma-like lesions. In addition, RT2;RVE mice exhibit activated lymphangiogenesis at the tumor periphery and increased neutrophil and macrophage tumor infiltration and micro-metastasis to lymph nodes and lungs. These phenotypes markedly differ from the phenotypes observed with the transgenic expression of the other VEGF family members in ß-cells of normal mice and of RT2 mice.


Asunto(s)
Hemangioma/etiología , Neoplasias Pancreáticas/etiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Virales/metabolismo , Animales , Proliferación Celular , Células Endoteliales/metabolismo , Células Endoteliales/patología , Cabras , Hemangioma/metabolismo , Hemangioma/patología , Humanos , Células Secretoras de Insulina/metabolismo , Linfangiogénesis/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ovinos , Transducción de Señal , Proteínas Virales/genética
5.
Clin Cancer Res ; 21(21): 4856-67, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26206868

RESUMEN

PURPOSE: Pancreatic neuroendocrine tumors (PNET) represent a rare but challenging heterogeneous group of cancers with an increasing incidence over the last number of decades. Herein, we report an in-depth evaluation of the new antiangiogenic small-molecule tyrosine kinase inhibitor (TKI) nintedanib in the preclinical Rip1Tag2 transgenic mouse model of neuroendocrine carcinoma of the pancreas (insulinoma). EXPERIMENTAL DESIGN: We have assessed the antiangiogenic and antitumor activity of nintedanib, in comparison with other antiangiogenic TKI, by treating Rip1Tag2 transgenic mice with different treatment schedules complemented with histopathologic, cell biologic, and biochemical analyses. RESULTS: Prolonged nintedanib treatment of Rip1Tag2 mice has led to a strong suppression of angiogenesis, accompanied by a reduced tumor burden, which translated into a significant prolongation of survival. Despite nintedanib's inhibitory action on perivascular cells, the blood vessels remaining after therapy displayed a considerably mature phenotype with tight perivascular cell coverage and preserved perfusion. Nintedanib treatment did not increase local tumor invasiveness or metastasis to the liver and pancreatic lymph nodes--a phenomenon that has been observed with antiangiogenic treatments of Rip1Tag2 transgenic mice in other laboratories. In contrast with the strong reduction in blood microvessel densities, nintedanib did not have any impact on tumor lymphangiogenesis. CONCLUSIONS: Based on our findings, we propose the clinical evaluation of the antiangiogenic drug nintedanib as a new treatment modality for PNET patients, notably in a direct comparison with already established therapeutic regimens, such as sunitinib.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Antineoplásicos/farmacología , Carcinoma Neuroendocrino/genética , Indoles/farmacología , Insulina/genética , Neoplasias Pancreáticas/genética , Regiones Promotoras Genéticas , Animales , Apoptosis/efectos de los fármacos , Carcinoma Neuroendocrino/tratamiento farmacológico , Carcinoma Neuroendocrino/mortalidad , Carcinoma Neuroendocrino/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Fenotipo , Transducción de Señal/efectos de los fármacos , Carga Tumoral , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Angiogenesis ; 18(3): 327-45, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26021306

RESUMEN

Tumor growth depends on the formation of new blood vessels (tumor angiogenesis) either from preexisting vessels or by the recruitment of bone marrow-derived cells. Despite encouraging results obtained with preclinical cancer models, the therapeutic targeting of tumor angiogenesis has thus far failed to deliver an enduring clinical response in cancer patients. One major obstacle for improving anti-angiogenic therapy is the lack of validated biomarkers, which allow patient stratification for suitable treatment and a rapid assessment of therapy response. Toward these goals, we have employed several mouse models of tumor angiogenesis to identify cell populations circulating in their blood that correlated with the extent of tumor angiogenesis and therapy response. Flow cytometry analyses of different combinations of cell surface markers that define subsets of bone marrow-derived cells were performed on peripheral blood mononuclear cells from tumor-bearing and healthy mice. We identified one cell population, CD45(dim)VEGFR1(-)CD31(low), that was increased in levels during active tumor angiogenesis in a variety of transgenic and syngeneic transplantation mouse models of cancer. Treatment with various anti-angiogenic drugs did not affect CD45(dim)VEGFR1(-)CD31(low) cells in healthy mice, whereas in tumor-bearing mice, a consistent reduction in their levels was observed. Gene expression profiling of CD45(dim)VEGFR1(-)CD31(low) cells characterized these cells as an immature B cell population. These immature B cells were then directly validated as surrogate marker for tumor angiogenesis and of pharmacologic responses to anti-angiogenic therapies in various mouse models of cancer.


Asunto(s)
Linfocitos B/inmunología , Biomarcadores/metabolismo , Neoplasias/patología , Neovascularización Patológica , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Membrana Celular/metabolismo , Técnicas de Cocultivo , Biología Computacional , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Antígenos Comunes de Leucocito/metabolismo , Leucocitos Mononucleares/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
FASEB J ; 29(8): 3411-25, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25911611

RESUMEN

De novo formation of blood vessels is a pivotal mechanism during cancer development. During the past few years, antiangiogenic drugs have been developed to target tumor vasculature. However, because of limitations and adverse effects observed with current therapies, there is a strong need for alternative antiangiogenic strategies. Using specific anti-junctional adhesion molecule (JAM)-B antibodies and Jam-b-deficient mice, we studied the role in antiangiogenesis of JAM-B. We found that antibodies against murine JAM-B, an endothelium-specific adhesion molecule, inhibited microvessel outgrowth from ex vivo aortic rings and in vitro endothelial network formation. In addition, anti-JAM-B antibodies blocked VEGF signaling, an essential pathway for angiogenesis. Moreover, increased aortic ring branching was observed in aortas isolated from Jam-b-deficient animals, suggesting that JAM-B negatively regulates proangiogenic pathways. In mice, JAM-B expression was detected in de novo-formed blood vessels of tumors, but anti-JAM-B antibodies unexpectedly did not reduce tumor growth. Accordingly, JAM-B deficiency in vivo had no impact on blood vessel formation, suggesting that targeting JAM-B in vivo may be offset by other proangiogenic mechanisms. In conclusion, despite the promising effects observed in vitro, targeting JAM-B during tumor progression seems to be inefficient as a stand-alone antiangiogenesis therapy.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Neovascularización Patológica/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Femenino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
8.
Cancer Lett ; 328(1): 18-26, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22922303

RESUMEN

Tie-1 and Tie-2 tyrosine kinase receptors are expressed specifically on vascular endothelial cells and on a certain subtype of macrophages implicated in angiogenesis, thus, they have been a major focus of angiogenesis research. Tie-1 and Tie-2 are essential for vascular maturation during developmental, physiological and pathological angiogenesis. Angiopoietin 1-4 (Ang-1-4) have been identified as bona fide ligands of the Tie-2 receptor, while Tie-1 remains an orphan receptor which is able to heterodimerize with Tie-2 and to modulate Tie-2 signal transduction. The most exhaustively studied angiopoietins are Ang-1 and Ang-2. Ang-1 is a critical player in vessel maturation and it mediates migration, adhesion and survival of endothelial cells. Ang-2 disrupts the connections between the endothelium and perivascular cells and promotes cell death and vascular regression. Yet, in conjunction with VEGF, Ang-2 promotes neo-vascularization. Hence, angiopoietins exert crucial roles in the angiogenic switch during tumor progression, and increased expression of Ang-2 relative to Ang-1 in tumors correlates with poor prognosis. Its central role in the regulation of physiological and pathological angiogenesis makes the angiopoietin/Tie signaling pathway a therapeutically attractive target for the treatment of vascular disease and cancer.


Asunto(s)
Angiopoyetinas/metabolismo , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Humanos , Receptor TIE-1/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal/fisiología
9.
Cancer Res ; 71(17): 5717-27, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21778249

RESUMEN

Members of the Angiopoietin family regulate various aspects of physiologic and pathologic angiogenesis. Although Angiopoietin-1 (Ang-1) decreases endothelial cell permeability and increases vascular stabilization via recruitment of pericytes and smooth muscle cells to growing blood vessels, Angiopoietin-2 (Ang-2) mediates angiogenic sprouting and vascular regression. In this study, we used the Rip1Tag2 transgenic mouse model of pancreatic ß-cell carcinogenesis to investigate the roles of Ang-1 and Ang-2 in tumor angiogenesis and tumor progression. On their own, transgenic expression of human Ang-1 or Ang-2 in pancreatic ß cells caused formation of peri-insular lymphatic vessels in the absence of effects on blood vessel density, islet morphology, or physiology. When crossed to Rip1Tag2 mice, both Ang-1-and Ang-2-expressing ß-cell tumors showed increased peritumoral lymphangiogenesis in the absence of metastasis to local lymph nodes or distant organs. There was no alteration in tumor outgrowth, blood vessel density, or vessel maturation in Ang-1-expressing tumors. In contrast, Ang-2-expressing tumors exhibited diminished pericyte recruitment to blood vessels that were dilated, nonfunctional, and highly permeable. These tumors were hemorrhagic, highly infiltrated by leukocytes, and impaired in outgrowth. Together, our findings establish that Ang-2 antagonizes Ang-1 function, leading to excessive vessel sprouting with impaired pericyte recruitment and vessel stabilization. The poor perfusion of immature blood vessels results in retarded tumor growth, defining an important pathophysiologic pathway required for efficient tumorigenesis.


Asunto(s)
Angiopoyetina 1/antagonistas & inhibidores , Angiopoyetina 2/metabolismo , Linfangiogénesis , Neovascularización Patológica/metabolismo , Neoplasias Pancreáticas/irrigación sanguínea , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Angiopoyetina 2/genética , Animales , Células Cultivadas , Humanos , Ratones , Ratones Transgénicos , Neovascularización Patológica/genética , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Cancer Res ; 71(11): 3781-91, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21507936

RESUMEN

Tumor-mobilized bone marrow-derived CD11b(+) myeloid cells promote tumor angiogenesis, but how and when these cells acquire proangiogenic properties is not fully elucidated. Here, we show that CD11b(+) myelomonocytic cells develop proangiogenic properties during their differentiation from CD34(+) hematopoietic progenitors and that placenta growth factor (PlGF) is critical in promoting this education. Cultures of human CD34(+) progenitors supplemented with conditioned medium from breast cancer cell lines or PlGF, but not from nontumorigenic breast epithelial lines, generate CD11b(+) cells capable of inducing endothelial cell sprouting in vitro and angiogenesis in vivo. An anti-Flt-1 mAb or soluble Flt-1 abolished the generation of proangiogenic activity during differentiation from progenitor cells. Moreover, inhibition of metalloproteinase activity, but not VEGF, during the endothelial sprouting assay blocked sprouting induced by these proangiogenic CD11b(+) myelomonocytes. In a mouse model of breast cancer, circulating CD11b(+) cells were proangiogenic in the sprouting assays. Silencing of PlGF in tumor cells prevented the generation of proangiogenic activity in circulating CD11b(+) cells, inhibited tumor blood flow, and slowed tumor growth. Peripheral blood of breast cancer patients at diagnosis, but not of healthy individuals, contained elevated levels of PlGF and circulating proangiogenic CD11b(+) myelomonocytes. Taken together, our results show that cancer cells can program proangiogenic activity in CD11b(+) myelomonocytes during differentiation of their progenitor cells in a PlGF-dependent manner. These findings impact breast cancer biology, detection, and treatment.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Antígeno CD11b/biosíntesis , Células Madre Hematopoyéticas/patología , Monocitos/patología , Proteínas Gestacionales/inmunología , Adulto , Anciano , Animales , Antígenos CD34/biosíntesis , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Femenino , Sangre Fetal/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Monocitos/inmunología , Factor de Crecimiento Placentario , Embarazo , Proteínas Gestacionales/farmacología , Proteínas Recombinantes/farmacología
11.
Expert Opin Ther Targets ; 13(1): 93-104, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19063709

RESUMEN

BACKGROUND: Metastatic melanoma remains one of the most aggressive forms of cancer, with a survival expectation of above six months only in rare cases. Despite advances in the characterization of the underlying molecular pathways and in the development of specific targeted treatments, available chemo- and immuno-therapy are unable to prolong survival significantly in advanced-stage melanoma. Rai like protein (RaLP) is a newly identified Src homology 2 domain containing (Shc) family member selectively expressed during the transition to metastatic melanoma and thus is a potential melanoma-specific drugable target. OBJECTIVE: To summarize progress in the ongoing therapeutic approaches to metastatic melanoma and discuss RaLP as a potential novel therapeutic target. METHODS: Current understanding of the major signaling pathways involved in melanoma metastatization and of the corresponding pharmacological inhibitors is discussed. CONCLUSION: RaLP might represent a new drugable target for the treatment of metastatic disease.


Asunto(s)
Melanoma/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Progresión de la Enfermedad , Humanos , Melanoma/patología , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Homología de Secuencia de Aminoácido , Proteínas Adaptadoras de la Señalización Shc/química
12.
Cancer Res ; 67(7): 3064-73, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17409413

RESUMEN

The Src homology and collagen (Src) family of adaptor proteins comprises six Shc-like proteins encoded by three loci in mammals (Shc, Rai, and Sli). Shc-like proteins are tyrosine kinase substrates, which regulate diverse signaling pathways and cellular functions, including Ras and proliferation (p52/p46Shc), phosphatidylinositol 3-kinase and survival (p54Rai), and mitochondrial permeability transition and apoptosis (p66Shc). Here, we report the identification, cloning, and sequence characterization of a new member of the Shc family that we termed RaLP. RaLP encodes a 69-kDa protein characterized by the CH2-PTB-CH1-SH2 modularity, typical of the Shc protein family, and expressed, among adult tissues, only in melanomas. Analysis of RaLP expression during the melanoma progression revealed low expression in normal melanocytes and benign nevi, whereas high levels of RaLP protein were found at the transition from radial growth phase to vertical growth phase and metastatic melanomas, when tumor cells acquire migratory competence and invasive potential. Notably, silencing of RaLP expression in metastatic melanomas by RNA interference reduced tumorigenesis in vivo. Analysis of RaLP in melanoma signal transduction pathways revealed that (a) when ectopically expressed in RaLP-negative melanocytes and nonmetastatic melanoma cells, it functions as a substrate of activated insulin-like growth factor-1 and epidermal growth factor receptors and increases Ras/mitogen-activated protein kinase (MAPK) signaling and cell migration, whereas (b) its silencing in RaLP-positive melanoma cells abrogates cell migration in vitro, without affecting MAPK signaling, suggesting that RaLP activates both Ras-dependent and Ras-independent migratory pathways in melanomas. These findings indicate that RaLP is a specific marker of metastatic melanomas, a critical determinant in the acquisition of the migratory phenotype by melanoma cells, and a potential target for novel anti-melanoma therapeutic strategies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular/fisiología , Melanoma/enzimología , Melanoma/patología , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencia de Aminoácidos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Clonación Molecular , Regulación hacia Abajo , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Sistema de Señalización de MAP Quinasas , Melanoma/genética , Melanoma/secundario , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor IGF Tipo 1/metabolismo , Proteínas Adaptadoras de la Señalización Shc , Transfección , Familia-src Quinasas/genética
13.
J Neurochem ; 91(4): 813-24, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15525335

RESUMEN

Oligodendrocytes and Schwann cells use diverse regulatory elements to transcribe myelin basic protein (Mbp). For example, an enhancer 9.0 kb upstream of Mbp (MbpSCE1) activated either the proximal Mbp or hsp68 promoters only in Schwann cells in transgenic mice. Here, we analyze MbpSCE1 in vitro and in vivo and show that MbpSCE1 also activates another myelin gene, Mpz, specifically in Schwann cells in transgenic mice. Surprisingly, although MbpSCE1 behaves as an enhancer in Schwann cells, we show here for the first time that it also activates transcription robustly in cultured oligodendrocytes, but only from its original genomic position. Diverse nuclear proteins binding at distinct combinations of DNA elements in the two cell types may account for cell-specific MbpSCE1 function. A further surprise is that MbpSCE1 activation does not depend on consensus binding sites for the myelin-associated PPARbeta or Krox 20 transcription factors. Finally, chromatin context augments activation of MbpSCE1. Therefore, MbpSCE1 is active in both Schwann cells and oligodendrocytes, but by diverse mechanisms.


Asunto(s)
Elementos de Facilitación Genéticos/fisiología , Proteína Básica de Mielina/genética , Células de Schwann/metabolismo , Animales , Células Cultivadas , Cromatina/metabolismo , Huella de ADN , Desoxirribonucleasa I , Ensayo de Cambio de Movilidad Electroforética , Elementos de Facilitación Genéticos/genética , Genes Reporteros , Humanos , Ratones , Ratones Transgénicos , Proteínas Nucleares/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Ratas , Ratas Sprague-Dawley , Células de Schwann/citología , Análisis de Secuencia de ADN , Eliminación de Secuencia , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA