Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Cancer ; 17(1): 485, 2017 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-28709411

RESUMEN

BACKGROUND: The serine/threonine protein kinases ROCK1 and 2 are key RhoA-mediated regulators of cell shape and cytoskeletal dynamics. These proteins perform multiple functions in vascular endothelial cell physiology and are attractive targets for cancer therapy based on their roles as oncogenes and metastatic promoters. Given their critical functions in both of these processes, we hypothesized that molecular targeting of ROCK proteins would be exceedingly effective against vascular tumors such as hemangiomas and angiosarcomas, which are neoplasms composed of aberrant endothelial cells. METHODS: In this study, we compared ROCK1 and 2 protein expression in a large panel of benign and malignant vascular tumors to that of normal vasculature. We then utilized shRNA technology to knockdown the expression of ROCK1 and 2 in SVR tumor-forming vascular cells, and evaluated tumor size and proliferation rate in a xenograft model. Finally, we employed proteomics and metabolomics to assess how knockdown of the ROCK paralogs induced alterations in protein expression/phosphorylation and metabolite concentrations in the xenograft tumors. RESULTS: Our findings revealed that ROCK1 was overexpressed in malignant vascular tumors such as hemangioendotheliomas and angiosarcomas, and ROCK2 was overexpressed in both benign and malignant vascular tumors including hemangiomas, hemangioendotheliomas, hemangiopericytomas, and angiosarcomas. shRNA-mediated knockdown of ROCK2, but not ROCK1, in xenograft vascular tumors significantly reduced tumor size and proliferative index compared to control tumors. Proteomics and metabolomics analysis of the xenograft tumors revealed both overlapping as well as unique roles for the ROCK paralogs in regulating signal transduction and metabolite concentrations. CONCLUSIONS: Collectively, these data indicate that ROCK proteins are overexpressed in diverse vascular tumors and suggest that specific targeting of ROCK2 proteins may show efficacy against malignant vascular tumors.


Asunto(s)
Neoplasias/genética , Neoplasias Vasculares/genética , Quinasas Asociadas a rho/genética , Animales , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Neoplasias/patología , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Activación Transcripcional/genética , Neoplasias Vasculares/clasificación , Neoplasias Vasculares/patología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Oncotarget ; 7(25): 38467-86, 2016 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-27509260

RESUMEN

Angiogenesis is hallmark of clear cell renal cell carcinogenesis. Anti-angiogenic therapies have been successful in improving disease outcome; however, most patients treated with anti-angiogenic agents will eventually progress. In this study we report that clear cell renal cell carcinoma was associated with vasculogenic mimicry in both mice and human with tumor cells expressing endothelial markers in the vicinity of tumor vessels. We show that vasculogenic mimicry was efficiently targeted by sunitinib but eventually associated with tumor resistance and a more aggressive phenotype both in vitro and in vivo. Re-challenging these resistant tumors in mice, we showed that second-line treatment with everolimus particularly affected vasculogenic mimicry and tumor cell differentiation compared to sorafenib and axitinib. Finally, our results highlighted the phenotypic and genotypic changes at the tumor cell and microenvironment levels during sunitinib response and progression and the subsequent improvement second-line therapies bring to the current renal cell carcinoma treatment paradigm.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Everolimus/farmacología , Indoles/farmacología , Neoplasias Renales/tratamiento farmacológico , Pirroles/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , Axitinib , Carcinoma de Células Renales/irrigación sanguínea , Carcinoma de Células Renales/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Imidazoles/farmacología , Indazoles/farmacología , Neoplasias Renales/irrigación sanguínea , Neoplasias Renales/patología , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Distribución Aleatoria , Sunitinib , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Am J Pathol ; 185(3): 717-28, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25622542

RESUMEN

Liposarcomas, which are malignant fatty tumors, are the second most common soft-tissue sarcomas. Several histologically defined liposarcoma subtypes exist, yet little is known about the molecular pathology that drives the diversity in these tumors. We used functional genomics to classify a panel of diverse liposarcoma cell lines based on hierarchical clustering of their gene expression profiles, indicating that liposarcoma gene expression profiles and histologic classification are not directly correlated. Boolean probability approaches based on cancer-associated properties identified differential expression in multiple genes, including MYC, as potentially affecting liposarcoma signaling networks and cancer outcome. We confirmed our method with a large panel of lipomatous tumors, revealing that MYC protein expression is correlated with patient survival. These data encourage increased reliance on genomic features in conjunction with histologic features for liposarcoma clinical characterization and lay the groundwork for using Boolean-based probabilities to identify prognostic biomarkers for clinical outcome in tumor patients.


Asunto(s)
Liposarcoma/genética , Proteínas Proto-Oncogénicas c-myc/genética , Neoplasias de los Tejidos Blandos/genética , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Femenino , Genómica , Humanos , Liposarcoma/mortalidad , Liposarcoma/patología , Masculino , Persona de Mediana Edad , Pronóstico , Neoplasias de los Tejidos Blandos/mortalidad , Neoplasias de los Tejidos Blandos/patología , Tasa de Supervivencia , Transcriptoma
4.
Nat Med ; 20(4): 408-14, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24658078

RESUMEN

Erythropoietin (EPO) stimulates proliferation of early-stage erythrocyte precursors and is widely used for the treatment of chronic anemia. However, several types of EPO-resistant anemia are characterized by defects in late-stage erythropoiesis, which is EPO independent. Here we investigated regulation of erythropoiesis using a ligand-trapping fusion protein (ACE-536) containing the extracellular domain of human activin receptor type IIB (ActRIIB) modified to reduce activin binding. ACE-536, or its mouse version RAP-536, produced rapid and robust increases in erythrocyte numbers in multiple species under basal conditions and reduced or prevented anemia in murine models. Unlike EPO, RAP-536 promoted maturation of late-stage erythroid precursors in vivo. Cotreatment with ACE-536 and EPO produced a synergistic erythropoietic response. ACE-536 bound growth differentiation factor-11 (GDF11) and potently inhibited GDF11-mediated Smad2/3 signaling. GDF11 inhibited erythroid maturation in mice in vivo and ex vivo. Expression of GDF11 and ActRIIB in erythroid precursors decreased progressively with maturation, suggesting an inhibitory role for GDF11 in late-stage erythroid differentiation. RAP-536 treatment also reduced Smad2/3 activation, anemia, erythroid hyperplasia and ineffective erythropoiesis in a mouse model of myelodysplastic syndromes (MDS). These findings implicate transforming growth factor-ß (TGF-ß) superfamily signaling in erythroid maturation and identify ACE-536 as a new potential treatment for anemia, including that caused by ineffective erythropoiesis.


Asunto(s)
Receptores de Activinas Tipo II , Anemia/sangre , Proteínas Morfogenéticas Óseas/efectos de los fármacos , Células Precursoras Eritroides/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Factores de Diferenciación de Crecimiento/efectos de los fármacos , Hematínicos/farmacología , Síndromes Mielodisplásicos/sangre , Proteínas Recombinantes de Fusión/farmacología , Animales , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Modelos Animales de Enfermedad , Quimioterapia Combinada , Recuento de Eritrocitos , Eritropoyetina/farmacología , Factores de Diferenciación de Crecimiento/antagonistas & inhibidores , Haplorrinos , Humanos , Ligandos , Ratones , Ratas , Recuento de Reticulocitos , Transducción de Señal/efectos de los fármacos , Proteína Smad2/efectos de los fármacos , Proteína smad3/efectos de los fármacos
5.
BMC Cancer ; 14: 81, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24511912

RESUMEN

BACKGROUND: Preclinical and clinical studies have shown for decades that tumor cells demonstrate significantly enhanced sensitivity to "fever range" hyperthermia (increasing the intratumoral temperature to 42-45°C) than normal cells, although it is unknown why cancer cells exhibit this distinctive susceptibility. METHODS: To address this issue, mammary epithelial cells and three malignant breast cancer lines were subjected to hyperthermic shock and microarray, bioinformatics, and network analysis of the global transcription changes was subsequently performed. RESULTS: Bioinformatics analysis differentiated the gene expression patterns that distinguish the heat shock response of normal cells from malignant breast cancer cells, revealing that the gene expression profiles of mammary epithelial cells are completely distinct from malignant breast cancer lines following this treatment. Using gene network analysis, we identified altered expression of transcripts involved in mitotic regulators, histones, and non-protein coding RNAs as the significant processes that differed between the hyperthermic response of mammary epithelial cells and breast cancer cells. We confirmed our data via qPCR and flow cytometric analysis to demonstrate that hyperthermia specifically disrupts the expression of key mitotic regulators and G2/M phase progression in the breast cancer cells. CONCLUSION: These data have identified molecular mechanisms by which breast cancer lines may exhibit enhanced susceptibility to hyperthermic shock.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Genómica/métodos , Calor , Hipertermia Inducida/métodos , Línea Celular Tumoral , Susceptibilidad a Enfermedades/diagnóstico , Femenino , Fiebre , Redes Reguladoras de Genes/genética , Humanos , Células MCF-7 , Análisis por Matrices de Proteínas
6.
Cancer Genomics Proteomics ; 10(6): 251-63, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24336634

RESUMEN

BACKGROUND: Therapeutic targeting of Rho-Associated, Coiled-Coil Containing Protein Kinase (ROCK) signaling for tumor cells and tumor endothelium has shown efficacy in pre-clinical tumors models, and a better understanding of how proteins regulate tumor progression will strengthen our knowledge over disease etiology and treatment of patients with cancer. Recent reports have shown that ROCK activity is critical for the expression of a large number of mRNA transcripts across multiple cell types including endothelial cells. MATERIALS AND METHODS: To examine the effects of ROCK proteins on microRNA (miRNA) expression in tumor-forming endothelial cells, we utilized microarrays to evaluate expression levels of 1088 miRNAs in vascular tumor-forming endothelial cells knocked-down for ROCK1 or ROCK2 or treated with a pharmacological inhibitor of ROCK activity. RESULTS: Microarray analysis demonstrated that inhibiting ROCK activity altered global miRNA expression. We confirmed our findings using qPCR and identified cell-cycle progression, calcium transport, and neurogenesis/synaptogenesis as the most highly overrepresented predicted target gene networks for the identified miRNAs whose expression was altered by ROCK inhibition. CONCLUSION: ROCK signaling induces large-scale changes in global miRNA expression and may lead to a better understanding of how these proteins affect aberrant vascular states.


Asunto(s)
Células Endoteliales/metabolismo , MicroARNs/biosíntesis , Quinasas Asociadas a rho/metabolismo , Animales , Línea Celular , Células Endoteliales/enzimología , Técnicas de Silenciamiento del Gen , Ratones , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Quinasas Asociadas a rho/deficiencia , Quinasas Asociadas a rho/genética
7.
FEBS J ; 280(18): 4474-94, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23802622

RESUMEN

Alterations in cell shape have been shown to modulate chromatin condensation and cell lineage specification; however, the mechanisms controlling these processes are largely unknown. Because endothelial cells experience cyclic mechanical changes from blood flow during normal physiological processes and disrupted mechanical changes as a result of abnormal blood flow, cell shape deformation and loss of polarization during coronary artery disease, we aimed to determine how morphological restriction affects global gene expression patterns. Human coronary artery endothelial cells (HCAECs) were cultured on spatially defined adhesive micropatterns, forcing them to conform to unique cellular morphologies differing in cellular polarization and angularity. We utilized pattern recognition algorithms and statistical analysis to validate the cytoskeletal pattern reproducibility and uniqueness of each micropattern, and performed microarray analysis on normal-shaped and micropatterned HCAECs to determine how constrained cellular morphology affects gene expression patterns. Analysis of the data revealed that forcing HCAECs to conform to geometrically-defined shapes significantly affects their global transcription patterns compared to nonrestricted shapes. Interestingly, gene expression patterns were altered in response to morphological restriction in general, although they were consistent regardless of the particular shape the cells conformed to. These data suggest that the ability of HCAECs to spread, although not necessarily their particular morphology, dictates their genomics patterns.


Asunto(s)
Citoesqueleto de Actina/genética , Vasos Coronarios/citología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Proteínas de Microfilamentos/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Algoritmos , Adhesión Celular , Polaridad Celular/genética , Forma de la Célula/genética , Vasos Coronarios/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Perfilación de la Expresión Génica , Humanos , Procesamiento de Imagen Asistido por Computador , Mecanotransducción Celular , Proteínas de Microfilamentos/metabolismo , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos , Cultivo Primario de Células
8.
PLoS One ; 8(3): e60021, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23555867

RESUMEN

Therapeutic targeting of the beta-adrenergic receptors has recently shown remarkable efficacy in the treatment of benign vascular tumors such as infantile hemangiomas. As infantile hemangiomas are reported to express high levels of beta adrenergic receptors, we examined the expression of these receptors on more aggressive vascular tumors such as hemangioendotheliomas and angiosarcomas, revealing beta 1, 2, and 3 receptors were indeed present and therefore aggressive vascular tumors may similarly show increased susceptibility to the inhibitory effects of beta blockade. Using a panel of hemangioendothelioma and angiosarcoma cell lines, we demonstrate that beta adrenergic inhibition blocks cell proliferation and induces apoptosis in a dose dependent manner. Beta blockade is selective for vascular tumor cells over normal endothelial cells and synergistically effective when combined with standard chemotherapeutic or cytotoxic agents. We demonstrate that inhibition of beta adrenergic signaling induces large scale changes in the global gene expression patterns of vascular tumors, including alterations in the expression of established cell cycle and apoptotic regulators. Using in vivo tumor models we demonstrate that beta blockade shows remarkable efficacy as a single agent in reducing the growth of angiosarcoma tumors. In summary, these experiments demonstrate the selective cytotoxicity and tumor suppressive ability of beta adrenergic inhibition on malignant vascular tumors and have laid the groundwork for a promising treatment of angiosarcomas in humans.


Asunto(s)
Hemangioendotelioma/tratamiento farmacológico , Hemangioendotelioma/metabolismo , Hemangiosarcoma/tratamiento farmacológico , Propranolol/uso terapéutico , Receptores Adrenérgicos beta/metabolismo , Antagonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/uso terapéutico , Animales , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citoesqueleto/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Hemangiosarcoma/metabolismo , Humanos , Inmunohistoquímica , Ratones , Propranolol/farmacología
9.
Vasc Cell ; 5(1): 6, 2013 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-23531100

RESUMEN

BACKGROUND: Infantile hemangiomas are benign vascular tumors primarily found on the skin in 10% of the pediatric population. The etiology of this disease is largely unknown and while large scale genomic studies have examined the transcriptomes of infantile hemangioma tumors as a whole, no study to date has compared the global gene expression profiles of pure infantile hemangioma endothelial cells (HEMECs) to that of normal human dermal microvascular endothelial cells (HDMVECs). METHODS: To shed light on the molecular differences between these normal and aberrant dermal endothelial cell types, we performed whole genome microarray analysis on purified cultures of HEMECs and HDMVECs. We then utilized qPCR and immunohistochemistry to confirm our microarray results. RESULTS: Our array analysis identified 125 genes whose expression was upregulated and 104 genes whose expression was downregulated by greater than two fold in HEMECs compared to HDMVECs. Bioinformatics analysis revealed three major classifications of gene functions that were altered in HEMECs including cell adhesion, cell cycle, and arachidonic acid production. Several of these genes have been reported to be critical regulators and/or mutated in cancer, vascular tumors, and vascular malformations. We confirmed the expression of a subset of these differentially expressed genes (ANGPT2, ANTXR1, SMARCE1, RGS5, CTAG2, LTBP2, CLDN11, and KISS1) using qPCR and utilized immunohistochemistry on a panel of paraffin embedded infantile hemangioma tumor tissues to demonstrate that the cancer/testis antigen CTAG2 is highly abundant in vessel-dense proliferating infantile hemangiomas and with significantly reduced levels during tumor involution as vascular density decreases. CONCLUSION: Our data reveal that the transcriptome of HEMECs is reflective of a pro-proliferative cell type with altered adhesive characteristics. Moveover, HEMECs show altered expression of many genes that are important in the progression and prognosis of metastatic cancers.

10.
Exp Ther Med ; 4(4): 594-604, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23170111

RESUMEN

Infantile hemangiomas (IHs) are non-malignant, largely cutaneous vascular tumors affecting approximately 5-10% of children to varying degrees. During the first year of life, these tumors are strongly proliferative, reaching an average size ranging from 2 to 20 cm. These lesions subsequently stabilize, undergo a spontaneous slow involution and are fully regressed by 5 to 10 years of age. Systemic treatment of infants with the non-selective ß-adrenergic receptor blocker, propranolol, has demonstrated remarkable efficacy in reducing the size and appearance of IHs. However, the mechanism by which this occurs is largely unknown. In this study, we sought to understand the molecular mechanisms underlying the effectiveness of ß blocker treatment in IHs. Our data reveal that propranolol treatment of IH endothelial cells, as well as a panel of normal primary endothelial cells, blocks endothelial cell proliferation, migration, and formation of the actin cytoskeleton coincident with alterations in vascular endothelial growth factor receptor-2 (VEGFR-2), p38 and cofilin signaling. Moreover, propranolol induces major alterations in the protein levels of key cyclins and cyclin-dependent kinase inhibitors, and modulates global gene expression patterns with a particular affect on genes involved in lipid/sterol metabolism, cell cycle regulation, angiogenesis and ubiquitination. Interestingly, the effects of propranolol were endothelial cell-type independent, affecting the properties of IH endothelial cells at similar levels to that observed in neonatal dermal microvascular and coronary artery endothelial cells. This data suggests that while propranolol markedly inhibits hemangioma and normal endothelial cell function, its lack of endothelial cell specificity hints that the efficacy of this drug in the treatment of IHs may be more complex than simply blockage of endothelial function as previously believed.

11.
J Cell Biochem ; 111(3): 543-53, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20626031

RESUMEN

Healthy cells, as well as benign and malignant tumors, depend upon the body's blood supply to bring in oxygen and nutrients and carry away waste products. Using this property against tumors, anti-angiogenic therapy targets the tumor vasculature with the aim of starving the tumor, and has demonstrated exceptional clinical efficacy against a number of tumors. This review discusses the current state of knowledge regarding anti-angiogenic therapies presently available to patients, and garners from both preclinical and clinical literature the benefits and side effects associated with anti-angiogenic therapies, the unfortunate mechanisms of acquired resistance to these novel therapeutics, and highlights promising next generation anti-angiogenics that may overcome the limitations encountered with first generation therapies.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Resistencia a Antineoplásicos , Humanos , Neoplasias/irrigación sanguínea
12.
FASEB J ; 24(9): 3186-95, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20400538

RESUMEN

The small GTPase RhoA and its downstream effectors, ROCK1 and ROCK2, regulate a number of cellular processes, including cell motility, proliferation, survival, and permeability. Pharmacological inhibitors of the Rho pathway reportedly block angiogenesis; however, the molecular details of this inhibition are largely unknown. We demonstrate that vascular endothelial growth factor-A (VEGF) rapidly induces RhoA activation in endothelial cells (ECs). Moreover, the pharmacological inhibition of ROCK1/2 using 10 microM Y-27632 (the IC(50) for this compound in ECs) strongly disrupts vasculogenesis in pluripotent embryonic stem cell cultures, VEGF-mediated regenerative angiogenesis in ex vivo retinal explants, and VEGF-mediated in vitro EC tube formation. Furthermore, using small interfering RNA knockdown and mouse heterozygote knockouts of ROCK1 and ROCK2, we provide data indicating that VEGF-driven angiogenesis is largely mediated through ROCK2. These data demonstrate that Rho/ROCK signaling is an important mediator in a number of angiogenic processes, including EC migration, survival, and cell permeability, and suggest that Rho/ROCK inhibition may prove useful for the treatment of angiogenesis-related disorders.


Asunto(s)
Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/farmacología , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Amidas/farmacología , Animales , Apoptosis , Western Blotting , Bovinos , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Humanos , Ratones , Microscopía Fluorescente , Piridinas/farmacología , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Quinasas Asociadas a rho/genética
13.
Mol Cancer Ther ; 9(2): 379-88, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20124460

RESUMEN

Activin receptor-like kinase-1 (ALK1) is a type I, endothelial cell-specific member of the transforming growth factor-beta superfamily of receptors known to play an essential role in modulating angiogenesis and vessel maintenance. In the present study, we sought to examine the angiogenic and tumorigenic effects mediated upon the inhibition of ALK1 signaling using a soluble chimeric protein (ALK1-Fc). Of 29 transforming growth factor-beta-related ligands screened by surface plasmon resonance, only bone morphogenetic protein (BMP9) and BMP10 displayed high-affinity binding to ALK1-Fc. In cell-based assays, ALK1-Fc inhibited BMP9-mediated Id-1 expression in human umbilical vein endothelial cells and inhibited cord formation by these cells on a Matrigel substrate. In a chick chorioallantoic membrane assay, ALK1-Fc reduced vascular endothelial growth factor-, fibroblast growth factor-, and BMP10-mediated vessel formation. The growth of B16 melanoma explants was also inhibited significantly by ALK1-Fc in this assay. Finally, ALK1-Fc treatment reduced tumor burden in mice receiving orthotopic grafts of MCF7 mammary adenocarcinoma cells. These data show the efficacy of chimeric ALK1-Fc proteins in mitigating vessel formation and support the view that ALK1-Fc is a powerful antiangiogenic agent capable of blocking vascularization.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Fragmentos Fc de Inmunoglobulinas/metabolismo , Neoplasias/metabolismo , Neovascularización Patológica , Proteínas Recombinantes de Fusión/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Células CHO , Cricetinae , Cricetulus , Células Endoteliales/citología , Endotelio Vascular/citología , Factor 2 de Diferenciación de Crecimiento/metabolismo , Humanos , Ratones , Resonancia por Plasmón de Superficie , Telangiectasia Hemorrágica Hereditaria/metabolismo
14.
J Foot Ankle Surg ; 47(2): 163-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18312925

RESUMEN

Metastatic lesions localized to the foot are rare. When present, such lesions are typically associated with a poor prognosis. A good history can help guide the clinician when formulating differential diagnoses for a questionable clinical presentation. We report the case of a patient presenting with findings indicative of a metatarsal stress fracture and an ingrown toenail, which eventually resulted in the diagnosis of metastatic disease from the lung. ACFAS Level of Clinical Evidence: 4.


Asunto(s)
Pie/patología , Fracturas por Estrés/diagnóstico , Neoplasias Pulmonares/diagnóstico , Huesos Metatarsianos/lesiones , Uñas Encarnadas/diagnóstico , Neoplasias Cutáneas/secundario , Dedos del Pie/patología , Anciano , Resultado Fatal , Humanos , Neoplasias Pulmonares/patología , Masculino , Huesos Metatarsianos/cirugía
15.
Mol Biol Cell ; 19(3): 994-1006, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18094043

RESUMEN

Angiogenesis is largely controlled by hypoxia-driven transcriptional up-regulation and secretion of vascular endothelial growth factor (VEGF) and its binding to the endothelial cell tyrosine receptor kinases, VEGFR1 and VEGFR2. Recent expression analysis suggests that VEGF is expressed in a cell-specific manner in normoxic adult tissue; however, the transcriptional regulation and role of VEGF in these tissues remains fundamentally unknown. In this report we demonstrate that VEGF is coordinately up-regulated during terminal skeletal muscle differentiation. We reveal that this regulation is mediated in part by MyoD homo- and hetero-dimeric transcriptional mechanisms. Serial deletions of the VEGF promoter elucidated a region containing three tandem CANNTG consensus MyoD sites serving as essential sites of direct interaction for MyoD-mediated up-regulation of VEGF transcription. VEGF-null embryonic stem (ES) cells exhibited reduced myogenic differentiation compared with wild-type ES cells, suggesting that VEGF may serve a role in skeletal muscle differentiation. We demonstrate that VEGFR1 and VEGFR2 are expressed at low levels in myogenic precursor cells and are robustly activated upon VEGF stimulation and that their expression is coordinately regulated during skeletal muscle differentiation. VEGF stimulation of differentiating C2C12 cells promoted myotube hypertrophy and increased myogenic differentiation, whereas addition of sFlt1, a VEGF inhibitor, resulted in myotube hypotrophy and inhibited myogenic differentiation. We further provide evidence indicating VEGF-mediated myogenic marker expression, mitogenic activity, migration, and prosurvival functions may contribute to increased myogenesis. These data suggest a novel mechanism whereby VEGF is coordinately regulated as part of the myogenic differentiation program and serves an autocrine function regulating skeletal myogenesis.


Asunto(s)
Desarrollo de Músculos , Músculo Esquelético/embriología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Activación Enzimática/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Desarrollo de Músculos/efectos de los fármacos , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Proteína MioD/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología
16.
Mol Vis ; 13: 1144-53, 2007 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-17653061

RESUMEN

PURPOSE: The small GTPases function as "molecular switches" by binding and releasing GTP to mediate downstream signaling effects. The Rho-family of GTPases is central in modulating cell differentiation and cytoskeletal changes. Since eye development requires comprehensive morphogenetic movements and extensive cellular differentiation, we hypothesize that different small GTPases may play important roles during morphogenesis of eye development. To explore this possibility, we examined the expression patterns of three major Rho-GTPases: RhoA, Rac1, and Cdc42 in embryonic, postnatal (one day after birth), and adult (two-month old) mouse eye. METHODS: Various ocular tissues were collected from embryonic, postnatal, and adult C57BL/6 mice. Western blots were conducted using total proteins extracted from cornea, retina, lens epithelial cells, and lens fiber cells of the adult mice or different fractions of rat lenses. Immunohistochemistry (IHC) was performed with 6 mum thick sections cut through the eye ball region of 11.5 pc, 14.5 pc, 17.5 pc, postnatal, and adult mice. Parallel controls were run using the rabbit preimmune and GTPase-specific antibodies blocked with saturating levels of corresponding peptide antigen. RESULTS: In the embryonic mouse eye, RhoA and Cdc42 expressions were initially detectable in all three compartments at 11.5 pc. However, Rac1 became easily detectable in these compartments at 14.5 pc. Increased levels of RhoA, Rac1, and Cdc42 were detected in the three compartments at 17.5 pc and the strongest signals for RhoA, Rac1, and Cdc42 were observed in the primary lens fiber cells at 17.5 pc. In the postnatal mouse eye, the three small GTPases were significantly expressed in both endothelial and epithelial cells of mouse cornea, epithelial cells of the ocular lens, photoreceptors, horizontal/amacrine/Muller's cells, and some ganglian cells of the retina. Much lower level of expression was observed in the corneal stroma fibroblasts, lens fiber cells, and the inner and outer plexiform layers of the mouse retina. In the adult mouse eye, all three Rho-GTPases were expressed in corneal epithelial cells and retina. However, only RhoA protein was detected in corneal endothelial cells and Rac1 protein detected in the ocular lens. CONCLUSIONS: The strong expression of the three small GTPases in the cornea, lens, and retina of mouse eye at embryonic 17.5 pc and postnatal stage suggests their important functions for the morphogenesis of the different compartments of the mouse eye. Particularly, high levels of expression of RhoA, Rac1, and Cdc42 in embryonic lens fiber cells suggest their involvement in differentiation of primary lens fiber cells. In the adult mouse eye, all three Rho-GTPases seem to be involved in differentiation of corneal epithelial cells and retina, however, RhoA alone may be required for endothelial cell differentiation and Rac1 likely plays an important role in supporting continuous lens growth and maintenance of lens transparency.


Asunto(s)
Envejecimiento/metabolismo , Ojo/embriología , Ojo/enzimología , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Animales Recién Nacidos , Córnea/embriología , Córnea/metabolismo , Embrión de Mamíferos/enzimología , Ojo/crecimiento & desarrollo , Inmunohistoquímica , Cristalino/embriología , Cristalino/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Retina/embriología , Retina/metabolismo , Distribución Tisular
17.
Endocrinology ; 148(10): 4821-8, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17656465

RESUMEN

Kisspeptins are natural ligands of G protein-coupled receptor-54. Activation of KiSS1/G protein-coupled receptor-54 signaling pathways results in potent activation of the hypothalamus-pituitary-gonadal axis and initiates puberty. Recent data have shown that in female mice, KiSS1 is positively regulated by estradiol (E(2)) in the anteroventral periventricular nucleus, an important reproductive neuroendocrine brain region, but negatively regulated in the arcuate nucleus. However, little is known about the molecular mechanisms governing E(2)-modulated KiSS1 expression. Here, we demonstrate that the expression level of the KiSS1 gene was up-regulated with the administration of E(2) in estrogen receptor alpha (ERalpha)-positive hypothalamic GT1-7 cells. Using transient transfection of human KiSS1 gene promoter coupled to a luciferase reporter, E(2) increases promoter activity in the presence of ERalpha. Deletion analysis of KiSS1 promoter indicates that the E(2)-regulated increase in promoter activity depends on the Sp1 sites of the proximal promoter region. Using both EMSAs and chromatin immunoprecipitation analysis, we determined that both Sp1 and Sp3 proteins constitutively associate with the four putative Sp1 sites in vitro, whereas the association of ERalpha with the KiSS1 promoter is dependent on E(2) exposure. Sp1 and ERalpha form a complex in vivo to mediate the E(2)-induced activation of KiSS1 promoter. Interestingly, Sp1 transactivates KiSS1 promoter activity, whereas Sp3 functions as a transcriptional repressor. Together, these results demonstrate that E(2)-dependent transcriptional activation of KiSS1 gene is mediated by ERalpha through the interaction of Sp1/Sp3 proteins with the GC-rich motifs of KiSS1 promoter, providing a molecular mechanism of how steroid hormone feedback regulates KiSS1 expression.


Asunto(s)
Receptor alfa de Estrógeno/fisiología , Estrógenos/fisiología , Regulación de la Expresión Génica , Factor de Transcripción Sp1/fisiología , Factor de Transcripción Sp3/fisiología , Proteínas Supresoras de Tumor/genética , Línea Celular , Estradiol/farmacología , Humanos , Kisspeptinas , Luciferasas/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , Transfección , Regulación hacia Arriba
18.
J Biol Chem ; 281(1): 51-8, 2006 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-16260418

RESUMEN

KiSS-1 has been shown to function as a tumor metastasis suppressor gene and reduce the number of metastases in different cancers. The expression of KiSS-1 or KiSS1, like other tumor suppressor, is commonly reduced or completely ablated in a variety of cancers via an unknown mechanism. Here we show that the loss of KiSS-1 expression in highly metastatic breast cancer cell lines correlates directly with the expression levels of two transcription factors, activator protein-2alpha (AP-2alpha) and specificity protein 1 (Sp1), which synergistically activate the transcriptional regulation of KiSS-1 in breast cancer cells. Although the KiSS-1 promoter contains multiple AP-2alpha binding elements, AP-2alpha-mediated regulation occurs indirectly through Sp1 sites, as determined by deletion and mutation analysis. Overexpression of AP-2alpha into highly metastatic breast cell lines did not alter KiSS-1 promoter-driven luciferase gene activity. However, co-transfection of AP-2alpha wild-type or the dominant negative form of AP-2 lacking its C-terminal DNA-binding domain, AP-2B, together with Sp1, increased KiSS-1 promoter activity dramatically, suggesting that AP-2alpha regulation of KiSS-1 transcription does not require direct binding to the KiSS-1 promoter. Furthermore, we demonstrated that AP-2alpha directly interacted with Sp1 to form transcription complexes at two tandem Sp1-binding sites of the promoter to activate KiSS-1 transcription. Together, our results indicate that AP-2alpha and Sp1 are strong transcriptional regulators of KiSS-1 and that loss or decreased expression of AP-2alpha in breast cancer may account for the loss of tumor metastasis suppressor KiSS-1 expression and thus increased cancer metastasis.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/secundario , Proteínas/genética , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción AP-2/metabolismo , Neoplasias de la Mama/fisiopatología , Línea Celular Tumoral , Secuencia de Consenso , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Kisspeptinas , Regiones Promotoras Genéticas/fisiología , Activación Transcripcional/fisiología , Proteínas Supresoras de Tumor
19.
Mol Cell Biol ; 25(24): 11089-101, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16314529

RESUMEN

Rho family guanine nucleotide exchange factors (GEFs) regulate diverse cellular processes including cytoskeletal reorganization, cell adhesion, and differentiation via activation of the Rho GTPases. However, no studies have yet implicated Rho-GEFs as molecular regulators of the mesenchymal cell fate decisions which occur during development and repair of tissue damage. In this study, we demonstrate that the steady-state protein level of the Rho-specific GEF GEFT is modulated during skeletal muscle regeneration and that gene transfer of GEFT into cardiotoxin-injured mouse tibialis anterior muscle exerts a powerful promotion of skeletal muscle regeneration in vivo. In order to molecularly characterize this regenerative effect, we extrapolate the mechanism of action by examining the consequence of GEFT expression in multipotent cell lines capable of differentiating into a number of cell types, including muscle and adipocyte lineages. Our data demonstrate that endogenous GEFT is transcriptionally upregulated during myogenic differentiation and downregulated during adipogenic differentiation. Exogenous expression of GEFT promotes myogenesis of C2C12 cells via activation of RhoA, Rac1, and Cdc42 and their downstream effector proteins, while a dominant-negative mutant of GEFT inhibits this process. Moreover, we show that GEFT inhibits insulin-induced adipogenesis in 3T3L1 preadipocytes. In summary, we provide the first evidence that the Rho family signaling pathways act as potential regulators of skeletal muscle regeneration and provide the first reported molecular mechanism illustrating how a mammalian Rho family GEF controls this process by modulating mesenchymal cell fate decisions.


Asunto(s)
Adipogénesis , Factores de Intercambio de Guanina Nucleótido/metabolismo , Desarrollo de Músculos , Músculo Esquelético/fisiología , Regeneración , Células 3T3-L1 , Adipogénesis/genética , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Citoplasma/química , Activación Enzimática , Técnicas de Transferencia de Gen , Factores de Intercambio de Guanina Nucleótido/análisis , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Ratones , Desarrollo de Músculos/genética , Proteína MioD/metabolismo , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Regeneración/genética , Factores de Intercambio de Guanina Nucleótido Rho , Transducción de Señal , Transcripción Genética , Regulación hacia Arriba , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
20.
J Cell Biochem ; 96(5): 1034-48, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16149059

RESUMEN

PSGR is a newly identified human prostate tissue-specific gene belonging to the G-protein coupled receptor (GPCR) family. Overexpression of PSGR is associated with human prostate intraepithelial neoplasia (PIN) and prostate tumors, suggesting PSGR may play an important role in early prostate cancer development and progression. To understand the regulation of tissue-specific expression of human PSGR and its upregulation mechanism in prostate cancers, we characterized the promoter region of PSGR and analyzed the control mechanism for PSGR expression in human prostate tissues/cells. In this report, we demonstrate that two distinct promoters control the transcriptional regulation of PSGR in human prostate cells. The first promoter region includes exon 1 and a TATA box at -31 site. The minimal DNA sequence with promoter activity is about 123 bp upstream of exon 1. Exon 1 contains tissue specific regulatory activity for the first promoter of PSGR gene. The second promoter is located in the upstream region of exon 2, which is a TATA-less and non-GC-rich promoter. Primer extension and RNA protection assays (RPA) revealed that the transcription driven by the second promoter is initiated at the junction of intron and exon 2 within a cluster of nucleotides located about 250 bp upstream from the junction. Both promoters show prostate cell-specific characteristics in our luciferase assays in transfected cells. Furthermore, we investigated the regulation of the promoter activities of the PSGR gene by different growth factors and cytokines, and demonstrated that interleukin-6 (IL-6) activates the promoter activities of PSGR in human prostate cancer cells. These data suggest that two functional promoters regulate the transcriptional expression of PSGR in human prostate tissues and PSGR is a new target for IL-6 transcriptional regulation.


Asunto(s)
Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas , Neoplasias de la Próstata/metabolismo , Receptores Odorantes/biosíntesis , Receptores Odorantes/genética , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Línea Celular Tumoral , Cartilla de ADN/química , Progresión de la Enfermedad , Elementos de Facilitación Genéticos , Exones , Genoma , Células HeLa , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Intrones , Luciferasas/metabolismo , Masculino , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Neoplasias de la Próstata/patología , ARN/química , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Programas Informáticos , Factores de Tiempo , Transcripción Genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...