Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
J Cardiovasc Transl Res ; 10(4): 348-358, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28397162

RESUMEN

Angiogenic growth factor therapy for ischemic cardiovascular disease carries a risk of stimulating atherosclerotic plaque growth. We evaluated risk benefit ratio of sustained administration of recombinant human placental growth factor (rhPlGF)-2 in mice with advanced atherosclerosis and chronic ischemic cardiomyopathy. We maintained apolipoprotein E-deficient mice on a high cholesterol diet and induced myocardial infarction by transient ligation at 4 weeks. At 8 weeks, we assessed left ventricular (LV) function and randomized mice to receive rhPlGF-2 or vehicle (VEH) subcutaneously for 28 days. Administration of rhPlGF-2 significantly increased PlGF plasma levels without adverse hemodynamic or systemic inflammatory effects. RhPlGF-2 did not increase plaque area, composition, or vulnerability in the aortic arch. RhPlGF-2 significantly improved contractile function and reduced LV end-systolic and end-diastolic volume indices with a concomitant increase in capillary and arteriolar density in ischemic myocardium. RhPlGF-2 may represent a promising therapeutic strategy in chronic ischemic cardiomyopathy.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Aorta/efectos de los fármacos , Enfermedades de la Aorta/tratamiento farmacológico , Aterosclerosis/tratamiento farmacológico , Cardiomiopatías/tratamiento farmacológico , Infarto del Miocardio/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Factor de Crecimiento Placentario/administración & dosificación , Función Ventricular Izquierda/efectos de los fármacos , Inductores de la Angiogénesis/toxicidad , Animales , Aorta/patología , Aorta/fisiopatología , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/fisiopatología , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/fisiopatología , Colesterol en la Dieta , Enfermedad Crónica , Modelos Animales de Enfermedad , Infusiones Subcutáneas , Masculino , Ratones Noqueados para ApoE , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/fisiopatología , Factor de Crecimiento Placentario/toxicidad , Placa Aterosclerótica , Proteínas Recombinantes/administración & dosificación , Recuperación de la Función , Volumen Sistólico/efectos de los fármacos , Factores de Tiempo , Rigidez Vascular/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
2.
Int J Cardiol ; 203: 534-42, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26569359

RESUMEN

OBJECTIVES: We investigated whether sustained infusion of recombinant human placental growth factor-2 (rhPlGF-2) improves myocardial perfusion and left ventricular (LV) function in a porcine model of ischemic cardiomyopathy (ICM). METHODS: We induced myocardial ischemia using a flow-limiting stent in the LAD. Four weeks later, we randomized pigs with confirmed myocardial dysfunction to blinded rhPlGF-2 administration (PlGF2, 15 µg/kg/day, 14 days) or PBS (CON). At 8 weeks, we measured hemodynamics, contractile function and regional perfusion at rest and during stress using MRI and microspheres. We evaluated neovascularization post mortem. RESULTS: RhPlGF-2 administration increased PlGF serum levels more than 63-fold (83 3 ± 361 versus 11 ± 5 pg/ml CON, P<0.05) without adverse effects. After 4weeks, rhPlGF-2 significantly enhanced perfusion in the ischemic region at rest (0.83 ± 0.32 versus 0.58 ± 0.21 ml/min/g CON, P<0.05) and during hyperemia (1.50 ± 0.50 versus 1.02 ± 0.46 ml/min/g CON, P<0.05). Consequently, regional contractile function in rhPlGF-2-treated pigs improved at rest (37 ± 15% versus 23 ± 9% CON, P<0.05) and during high dose dobutamine stress (53 ± 31% versus 27 ± 16% CON, P<0.05). Enhanced perfusion translated into a greater improvement in LV ejection fraction and in preload-recruitable stroke work in rhPlGF-2-treated animals than in CON (52 ± 11 versus 41 ± 9%, and 76 ± 24 versus 41 ± 21 mmHg, respectively, P<0.05 for both), which was associated with significantly greater vascular density in the ischemic region. CONCLUSIONS: In chronic ICM, systemic rhPlGF-2 administration significantly enhances regional myocardial perfusion, contractile function at rest and during stress, and induces a prominent recovery of global cardiac function. PlGF-2 protein infusion is safe and may represent a promising therapy in chronic ICM.


Asunto(s)
Proteínas de la Membrana/uso terapéutico , Isquemia Miocárdica/tratamiento farmacológico , Guías de Práctica Clínica como Asunto , Proteínas Recombinantes/uso terapéutico , Animales , Enfermedad Crónica , Cricetinae , Modelos Animales de Enfermedad , Femenino , Estudios de Seguimiento , Masculino , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/metabolismo , Miocardio/patología , Porcinos , Factores de Tiempo , Resultado del Tratamiento
4.
Arterioscler Thromb Vasc Biol ; 31(12): 2881-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21940951

RESUMEN

OBJECTIVE: Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an adult-onset neurovascular disorder caused by stereotyped mutations in the NOTCH3 receptor. Elucidation of its pathobiology is still incomplete and remains a challenge, in part because the available preclinical mouse models to date do not reproduce the full spectrum of CADASIL pathology and clinical disease. METHODS AND RESULTS: Here, we report a novel knock-in mouse with Arg170Cys substitution in murine Notch3, corresponding to the prevalent Arg169Cys substitution in CADASIL. The Notch3(Arg170Cys) mice displayed late-onset, dominant CADASIL arteriopathy with typical granular osmiophilic material deposition and developed brain histopathology including thrombosis, microbleeds, gliosis, and microinfarction. Furthermore, Notch3(Arg170Cys) mice experienced neurological symptoms with motor defects such as staggering gait and limb paresis. CONCLUSIONS: This model, for the first time, phenocopies the arteriopathy and the histopathologic as well as clinical features of CADASIL and may offer novel opportunities to investigate disease pathogenesis.


Asunto(s)
Arginina , CADASIL/genética , CADASIL/patología , Cistina , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Receptores Notch/genética , Animales , Encéfalo/patología , CADASIL/fisiopatología , Capilares/patología , Fertilidad/fisiología , Ratones , Actividad Motora/fisiología , Músculo Liso Vascular/patología , Mutación/genética , Receptor Notch3
5.
J Am Coll Cardiol ; 55(20): 2232-43, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20466204

RESUMEN

OBJECTIVES: We compared biological repair after acute myocardial infarction (AMI) with selected porcine progenitor cell populations. BACKGROUND: Cell types and mechanisms responsible for myocardial repair after AMI remain uncertain. METHODS: In a blinded, randomized study, we infused autologous late-outgrowth endothelial progenitor cells (EPC) (n = 10, 34 +/- 22 x 10(6) CD29-31-positive, capable of tube formation), allogeneic green fluorescent peptide-labeled mesenchymal stem cells (MSC) (n = 11, 10 +/- 2 x 10(6) CD29-44-90-positive, capable of adipogenic and osteogenic differentiation), or vehicle (CON) (n = 12) in the circumflex artery 1 week after AMI. Systolic function (ejection fraction), left ventricular (LV) end-diastolic and end-systolic volumes, and infarct size were assessed with magnetic resonance imaging at 1 week and 7 weeks. Cell engraftment and vascular density were evaluated on postmortem sections. RESULTS: Recovery of LV ejection fraction from 1 to 7 weeks was similar between groups, but LV remodeling markedly differed with a greater increase of LV end-systolic volume in MSC and CON (+11 +/- 12 ml/m(2) and +7 +/- 8 ml/m(2) vs. -3 +/- 11 ml/m(2) in EPC, respectively, p = 0.04), and a similar trend was noted for LV end-diastolic volume (p = 0.09). After EPC, infarct size decreased more in segments with >50% infarct transmurality (p = 0.02 vs. MSC and CON) and was associated with a greater vascular density (p = 0.01). Late outgrowth EPCs secrete higher levels of the pro-angiogenic placental growth factor (733 [277 to 1,214] pg/10(6) vs. 59 [34 to 88] pg/10(6) cells in MSC, p = 0.03) and incorporate in neovessels in vivo. CONCLUSIONS: Infusion of late-outgrowth EPCs after AMI improves myocardial infarction remodeling via enhanced neovascularization but does not mediate cardiomyogenesis. Endothelial progenitor cell transfer might hold promise for heart failure prevention via pro-angiogenic or paracrine matrix-modulating effects.


Asunto(s)
Infarto del Miocardio/fisiopatología , Remodelación Ventricular/fisiología , Animales , Células Cultivadas , Inmunohistoquímica , Operón Lac/fisiología , Imagen por Resonancia Cinemagnética , Metaloproteinasas de la Matriz/sangre , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Reperfusión Miocárdica , Neovascularización Fisiológica/fisiología , Comunicación Paracrina/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante de Células Madre/métodos
6.
Cell ; 141(1): 178-90, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20371353

RESUMEN

Our findings that PlGF is a cancer target and anti-PlGF is useful for anticancer treatment have been challenged by Bais et al. Here we take advantage of carcinogen-induced and transgenic tumor models as well as ocular neovascularization to report further evidence in support of our original findings of PlGF as a promising target for anticancer therapies. We present evidence for the efficacy of additional anti-PlGF antibodies and their ability to phenocopy genetic deficiency or silencing of PlGF in cancer and ocular disease but also show that not all anti-PlGF antibodies are effective. We also provide additional evidence for the specificity of our anti-PlGF antibody and experiments to suggest that anti-PlGF treatment will not be effective for all tumors and why. Further, we show that PlGF blockage inhibits vessel abnormalization rather than density in certain tumors while enhancing VEGF-targeted inhibition in ocular disease. Our findings warrant further testing of anti-PlGF therapies.


Asunto(s)
Neovascularización Fisiológica/efectos de los fármacos , Proteínas Gestacionales/antagonistas & inhibidores , Proteínas Gestacionales/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Anticuerpos Monoclonales/uso terapéutico , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/prevención & control , Coroides/irrigación sanguínea , Modelos Animales de Enfermedad , Oftalmopatías/patología , Humanos , Neoplasias Hepáticas Experimentales/irrigación sanguínea , Neoplasias Hepáticas Experimentales/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Papiloma/irrigación sanguínea , Papiloma/inducido químicamente , Papiloma/prevención & control , Factor de Crecimiento Placentario , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/prevención & control
7.
Cardiovasc Res ; 86(1): 29-36, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19952000

RESUMEN

AIMS: Placental growth factor (PlGF), a homologue of vascular endothelial growth factor, is a pleiotropic cytokine with a pro-inflammatory activity. Previous gene-inactivation studies revealed that the loss of PlGF delays atherosclerotic lesion development and inhibits macrophage infiltration, but the activity of an anti-PlGF antibody (alphaPlGF mAb) has not been evaluated yet. METHODS AND RESULTS: We characterized the potential of short-term delivery of alphaPlGF mAb in inhibiting lesion development in ApoE-deficient mice (apoE(-/-)) and in CD4:TGFbetaRII(DN) x apoE(-/-) mice, a more severe atherosclerosis model. Short-term treatment of alphaPlGF mAb reduces early atherosclerotic plaque size and inflammatory cell infiltration in the lesion. CONCLUSION: These pharmacological alphaPlGF mAb results confirm previous genetic evidence that inhibition of PlGF slows down early atherosclerotic lesion development. Furthermore, the phenocopy of genetic and pharmacological loss-of-function strategies underscores that alphaPlGF acts by selectively neutralizing PlGF.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Enfermedades de la Aorta , Aterosclerosis , Proteínas Gestacionales/inmunología , Proteínas Gestacionales/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/tratamiento farmacológico , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Aterosclerosis/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Macrófagos/patología , Ratones , Ratones Mutantes , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Factor de Crecimiento Placentario , Túnica Íntima/metabolismo , Túnica Íntima/patología , Molécula 1 de Adhesión Celular Vascular/metabolismo
8.
Basic Res Cardiol ; 105(2): 169-79, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20020305

RESUMEN

Nitric oxide modulates the severity of myocardial ischemia-reperfusion (I/R) injury. We investigated whether cardioselective nitric oxide synthase 3 (NOS3) gene transfer could confer myocardial protection against I/R injury in pigs and examined potential molecular mechanisms. I/R injury was induced by balloon occlusion of the left anterior descending artery for 45 min followed by 4 or 72 h reperfusion. Hemodynamic and pathological changes were measured in pigs in the absence (n = 11) or presence of prior intracoronary retroinfusion of human NOS3 (AdNOS3, 5 x 10(10) PFU, n = 13) or control vector (AdRR5, 5 x 10(10) PFU, n = 11). Retrograde NOS3 gene transfer selectively increased NOS3 expression and NO bioavailability in the area at risk (AAR) without changing endogenous NOS isoform expression. At 4 h R, LV systolic (dP/dt(max)) and diastolic (dP/dt(min)) function was better preserved in AdNOS3- than in AdRR5-injected pigs (2,539 +/- 165 vs. 1,829 +/- 156 mmHg/s, and -2,781 +/- 340 vs. -2,062 +/- 292 mmHg/s, respectively, P < 0.05 for both). Myocardial infarct size (% AAR) was significantly smaller in AdNOS3 than in control and AdRR5 and associated with a significantly greater reduction in cardiac myeloperoxidase activity, a marker of neutrophil infiltration. The latter effects were sustained at 72 h R in a subset of pigs (n = 7). In the AAR, intercellular endothelial adhesion molecule-1 expression and cardiomyocyte apoptosis were significantly lower in AdNOS3. In conclusion, single myocardial NOS3 retroinfusion attenuates I/R injury, and causes a sustained reduction in myocardial infarct size and inflammatory cell infiltration. Gene-based strategies to increase NO bioavailability may have therapeutic potential in myocardial I/R.


Asunto(s)
Terapia Genética , Daño por Reperfusión Miocárdica/prevención & control , Óxido Nítrico Sintasa de Tipo III/genética , Animales , Apoptosis , Células Endoteliales/fisiología , Hemodinámica , Leucocitos/fisiología , Infarto del Miocardio/etiología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Distribución Aleatoria , Porcinos , Transgenes
9.
Drug Discov Today ; 13(17-18): 798-806, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18602017

RESUMEN

Thrombocytopenia is a common medical problem. The first generation thrombopoietic agents, recombinant THPO and 'megakaryocyte growth and development factor' (PEG-rHuMGDF) entered clinical trials, but their development was discontinued owing to neutralizing auto-antibodies cross-reacting with endogenous THPO, causing thrombocytopenia in healthy volunteers. Although an approved drug for prevention of severe thrombocytopenia following myelosuppressive chemotherapy (human Interleukin-11) exists, the search for new thrombopoietic agents continued because its use is limited by side effects. Several second generation thrombopoietic factors have entered clinical trials and some new negative regulators of megakaryopoiesis have been found, such as platelet factor 4 (PF4) and the pituitary adenylate cyclase activating polypeptide (PACAP). Their inhibition may be useful in the treatment of thrombocytopenia. This article reviews second generation thrombopoietic factors and those recently discovered regulators of megakaryopoiesis.


Asunto(s)
Plaquetas/efectos de los fármacos , Trombocitopenia/tratamiento farmacológico , Animales , Plaquetas/fisiología , Proliferación Celular/efectos de los fármacos , Humanos , Megacariocitos/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/uso terapéutico
10.
Stem Cells ; 26(4): 1017-26, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18258720

RESUMEN

Circulating endothelial progenitor cells (EPCs) contribute to neovascularization of ischemic tissues and repair of injured endothelium. The role of bone marrow-derived progenitor cells in hypoxia-induced pulmonary vascular remodeling and their tissue-engineering potential in pulmonary hypertension (PH) remain largely unknown. We studied endogenous mobilization and homing of EPCs in green fluorescent protein bone marrow chimeric mice exposed to chronic hypoxia, a common hallmark of PH. Despite increased peripheral mobilization, as shown by flow cytometry and EPC culture, bone marrow-derived endothelial cell recruitment in remodeling lung vessels was limited. Moreover, transfer of vascular endothelial growth factor receptor-2+/Sca-1+/CXCR-4+-cultured early-outgrowth EPCs failed to reverse PH, suggesting hypoxia-induced functional impairment of transferred EPCs. Chronic hypoxia decreased migration to stromal cell-derived factor-1alpha, adhesion to fibronectin, incorporation into a vascular network, and nitric oxide production (-41%, -29%, -30%, and -32%, respectively, vs. normoxic EPCs; p < .05 for all). The dysfunctional phenotype of hypoxic EPCs significantly impaired their neovascularization capacity in chronic hind limb ischemia, contrary to normoxic EPCs cultured in identical conditions. Mechanisms contributing to EPC dysfunction include reduced integrin alphav and beta1 expression, decreased mitochondrial membrane potential, and enhanced senescence. Novel insights from chronic hypoxia-induced EPC dysfunction may provide important cues for improved future cell repair strategies.


Asunto(s)
Células Endoteliales/fisiología , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Células Madre/patología , Animales , Células Cultivadas , Enfermedad Crónica , Células Endoteliales/patología , Células Endoteliales/trasplante , Miembro Posterior/irrigación sanguínea , Miembro Posterior/cirugía , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/cirugía , Hipoxia/patología , Hipoxia/cirugía , Ratones , Ratones Endogámicos C57BL , Células Madre/fisiología , Factores de Tiempo
11.
Neurosci Lett ; 432(1): 46-9, 2008 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-18164548

RESUMEN

Urokinase-type plasminogen activator receptor (uPAR) is a key component of the plasminogen activation system at the cell surface. Recent studies showed that uPAR is expressed in the ischemic damaged brain, suggesting its involvement in brain damage. In this study, we evaluated the role of uPAR in ischemic brain damage induced by permanent middle cerebral artery (MCA) occlusion in mice with genetic deficiency of uPAR (uPAR(-/-)) or of uPA (uPA(-/-)). Brain damage at 3 days was smaller in uPAR(-/-) mice (4.5+/-1.0mm(3)) than in littermate wild-type mice (uPAR(+/+)) (9.1+/-1.8mm(3), p<0.05), whereas it was comparable in uPA(-/-) (8.0+/-4.1mm(3)) and uPA(+/+) (6.9+/-2.6mm(3)) mice. uPAR expression was upregulated in the ipsilateral cerebral cortex within 12h, and remained elevated for up to 3 days. At 1 or 2 days after MCA occlusion, uPAR expression was selectively localized in vessels at the border of the damaged area. These findings suggest that uPAR expressed by endothelial cells augments the ischemic brain damage via a uPA-independent mechanism.


Asunto(s)
Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Activador de Plasminógeno de Tipo Uroquinasa/genética , Animales , Isquemia Encefálica/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Células Endoteliales/fisiología , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , ARN Mensajero/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
12.
J Clin Invest ; 118(2): 583-96, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18188450

RESUMEN

Many patients with anemia fail to respond to treatment with erythropoietin (Epo), a commonly used hormone that stimulates erythroid progenitor production and maturation by human BM or by murine spleen. The protein product of growth arrest-specific gene 6 (Gas6) is important for cell survival across several cell types, but its precise physiological role remains largely enigmatic. Here, we report that murine erythroblasts released Gas6 in response to Epo and that Gas6 enhanced Epo receptor signaling by activating the serine-threonine kinase Akt in these cells. In the absence of Gas6, erythroid progenitors and erythroblasts were hyporesponsive to the survival activity of Epo and failed to restore hematocrit levels in response to anemia. In addition, Gas6 may influence erythropoiesis via paracrine erythroblast-independent mechanisms involving macrophages. When mice with acute anemia were treated with Gas6, the protein normalized hematocrit levels without causing undesired erythrocytosis. In a transgenic mouse model of chronic anemia caused by insufficient Epo production, Gas6 synergized with Epo in restoring hematocrit levels. These findings may have implications for the treatment of patients with anemia who fail to adequately respond to Epo.


Asunto(s)
Anemia/tratamiento farmacológico , Anemia/genética , Eritropoyesis/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Animales , Adhesión Celular/genética , Supervivencia Celular , Modelos Animales de Enfermedad , Resistencia a Medicamentos , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Eritropoyetina/genética , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Mutantes , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Eritropoyetina/agonistas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
13.
Development ; 134(24): 4381-93, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18039968

RESUMEN

During vertebrate development, signaling by the TGFbeta ligand Nodal is critical for mesoderm formation, correct positioning of the anterior-posterior axis, normal anterior and midline patterning, and left-right asymmetric development of the heart and viscera. Stimulation of Alk4/EGF-CFC receptor complexes by Nodal activates Smad2/3, leading to left-sided expression of target genes that promote asymmetric placement of certain internal organs. We identified Ttrap as a novel Alk4- and Smad3-interacting protein that controls gastrulation movements and left-right axis determination in zebrafish. Morpholino-mediated Ttrap knockdown increases Smad3 activity, leading to ectopic expression of snail1a and apparent repression of e-cadherin, thereby perturbing cell movements during convergent extension, epiboly and node formation. Thus, although the role of Smad proteins in mediating Nodal signaling is well-documented, the functional characterization of Ttrap provides insight into a novel Smad partner that plays an essential role in the fine-tuning of this signal transduction cascade.


Asunto(s)
Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Receptores de Activinas/genética , Receptores de Activinas/metabolismo , Receptores de Activinas Tipo I , Animales , Secuencia de Bases , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Cadherinas/genética , Cadherinas/metabolismo , Cartilla de ADN/genética , Gastrulación/genética , Gastrulación/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteína Nodal , Oligorribonucleótidos Antisentido/genética , Transducción de Señal , Proteína smad3/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
14.
Cell ; 131(3): 463-75, 2007 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-17981115

RESUMEN

Novel antiangiogenic strategies with complementary mechanisms are needed to maximize efficacy and minimize resistance to current angiogenesis inhibitors. We explored the therapeutic potential and mechanisms of alphaPlGF, an antibody against placental growth factor (PlGF), a VEGF homolog, which regulates the angiogenic switch in disease, but not in health. alphaPlGF inhibited growth and metastasis of various tumors, including those resistant to VEGF(R) inhibitors (VEGF(R)Is), and enhanced the efficacy of chemotherapy and VEGF(R)Is. alphaPlGF inhibited angiogenesis, lymphangiogenesis, and tumor cell motility. Distinct from VEGF(R)Is, alphaPlGF prevented infiltration of angiogenic macrophages and severe tumor hypoxia, and thus, did not switch on the angiogenic rescue program responsible for resistance to VEGF(R)Is. Moreover, it did not cause or enhance VEGF(R)I-related side effects. The efficacy and safety of alphaPlGF, its pleiotropic and complementary mechanism to VEGF(R)Is, and the negligible induction of an angiogenic rescue program suggest that alphaPlGF may constitute a novel approach for cancer treatment.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/fisiología , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas Gestacionales/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/efectos adversos , Antineoplásicos/farmacología , Línea Celular , Movimiento Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Salud , Humanos , Linfangiogénesis/efectos de los fármacos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neovascularización Patológica/tratamiento farmacológico , Factor de Crecimiento Placentario , Resultado del Tratamiento
15.
Circulation ; 116(8): 936-43, 2007 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-17679618

RESUMEN

BACKGROUND: Nitric oxide (NO) activates soluble guanylate cyclase (sGC), a heterodimer composed of alpha- and beta-subunits, to produce cGMP. NO reduces pulmonary vascular remodeling, but the role of sGC in vascular responses to acute and chronic hypoxia remains incompletely elucidated. We therefore studied pulmonary vascular responses to acute and chronic hypoxia in wild-type (WT) mice and mice with a nonfunctional alpha1-subunit (sGCalpha1-/-). METHODS AND RESULTS: sGCalpha1-/- mice had significantly reduced lung sGC activity and vasodilator-stimulated phosphoprotein phosphorylation. Right ventricular systolic pressure did not differ between genotypes at baseline and increased similarly in WT (22+/-2 to 34+/-2 mm Hg) and sGCalpha1-/- (23+/-2 to 34+/-1 mm Hg) mice in response to acute hypoxia. Inhaled NO (40 ppm) blunted the increase in right ventricular systolic pressure in WT mice (22+/-2 to 24+/-2 mm Hg, P<0.01 versus hypoxia without NO) but not in sGCalpha1-/- mice (22+/-1 to 33+/-1 mm Hg) and was accompanied by a significant rise in lung cGMP content only in WT mice. In contrast, the NO-donor sodium nitroprusside (1.5 mg/kg) decreased systemic blood pressure similarly in awake WT and sGCalpha1-/- mice as measured by telemetry (-37+/-2 versus -42+/-4 mm Hg). After 3 weeks of hypoxia, the increases in right ventricular systolic pressure, right ventricular hypertrophy, and muscularization of intra-acinar pulmonary vessels were 43%, 135%, and 46% greater, respectively, in sGCalpha1-/- than in WT mice (P<0.01). Increased remodeling in sGCalpha1-/- mice was associated with an increased frequency of 5'-bromo-deoxyuridine-positive vessels after 1 and 3 weeks (P<0.01 versus WT). CONCLUSIONS: Deficiency of sGCalpha1 does not alter hypoxic pulmonary vasoconstriction. sGCalpha1 is essential for NO-mediated pulmonary vasodilation and limits chronic hypoxia-induced pulmonary vascular remodeling.


Asunto(s)
Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Vasodilatación/fisiología , Enfermedad Aguda , Animales , Antimetabolitos/farmacocinética , Presión Sanguínea/fisiología , Bromodesoxiuridina/farmacocinética , Enfermedad Crónica , GMP Cíclico/metabolismo , Dimerización , Femenino , Guanilato Ciclasa/química , Hipertensión Pulmonar/metabolismo , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/fisiopatología , Hipoxia/metabolismo , Masculino , Ratones , Ratones Mutantes , Arteria Pulmonar/fisiología , Circulación Pulmonar/fisiología , Receptores Citoplasmáticos y Nucleares/química , Guanilil Ciclasa Soluble , Función Ventricular Derecha/fisiología
16.
J Thromb Thrombolysis ; 24(1): 1-5, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17278000

RESUMEN

BACKGROUND: We investigated the safety and efficacy of several dosing regimens of catheter-directed staphylokinase (SY162) bolus administration for the treatment of long-term venous access catheter occlusion. METHODS: This open-label, ascending dose study enrolled 24 subjects. Three doses of SY162 were evaluated in three cohorts (0.15 mg, 0.3 mg and 0.45 mg) with eight subjects each. Catheter function was evaluated 30 min after the first bolus administration. In case of incomplete catheter function restoration, a second bolus was administered with reassessment of catheter function 30 min thereafter. Cathetergram was repeated to assess thrombus resolution. RESULTS: Complete restoration of catheter withdrawal function was observed in 2 (25%), 1 (13%) and 7 (88%) subjects after the first bolus in the first, second and third cohort respectively and in 4 (50%), 7 (88%) and 7 (88%) patients after the second administration of SY162. There were no bleeding complications nor other adverse events related to SY162. CONCLUSIONS: The doses tested in this trial were well tolerated and safe. A dose-response effect within the dose range tested was observed. A 2 ml bolus injection of 0.45 mg SY162 resulted in optimal efficacy after 30 min.


Asunto(s)
Catéteres de Permanencia/efectos adversos , Fibrinolíticos/uso terapéutico , Metaloendopeptidasas/uso terapéutico , Trombosis/prevención & control , Adulto , Anciano , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Humanos , Persona de Mediana Edad , Tiempo de Tromboplastina Parcial , Radiografía , Seguridad , Trombosis/diagnóstico por imagen
17.
Blood ; 109(11): 4742-52, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17299096

RESUMEN

We explored the physiologic role of endothelial cell apoptosis during development by generating mouse embryos lacking the inhibitor of apoptosis protein (IAP) survivin in endothelium. This was accomplished by intercrossing survivin(lox/lox) mice with mice expressing cre recombinase under the control of the endothelial cell specific tie1 promoter (tie1-cre mice). Lack of endothelial cell survivin resulted in embryonic lethality. Mutant embryos had prominent and diffuse hemorrhages from embryonic day 9.5 (E9.5) and died before E13.5. Heart development was strikingly abnormal. Survivin-null endocardial lineage cells could not support normal epithelial-mesenchymal transformation (EMT), resulting in hypoplastic endocardial cushions and in utero heart failure. In addition, 30% of mutant embryos had neural tube closure defects (NTDs) that were not caused by bleeding or growth retardation, but were likely due to alterations in the release of soluble factors from endothelial cells that otherwise support neural stem cell proliferation and neurulation. Thus, regulation of endothelial cell survival, and maintenance of vascular integrity by survivin are crucial for normal embryonic angiogenesis, cardiogenesis, and neurogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Miocardio/metabolismo , Neovascularización Patológica , Cresta Neural/citología , Defectos del Tubo Neural/genética , Animales , Apoptosis , Cruzamientos Genéticos , Medios de Cultivo Condicionados/farmacología , Modelos Animales de Enfermedad , Células Endoteliales/citología , Genotipo , Proteínas Inhibidoras de la Apoptosis , Ratones , Ratones Transgénicos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Proteínas Represoras , Survivin
18.
Thromb Haemost ; 96(6): 731-7, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17139366

RESUMEN

To investigate the effect of tiplaxtinin, designed as a synthetic inhibitor of plasminogen activator inhibitor-1 (PAI-1), on obesity, male C57Bl/6 mice (13-14 weeks old) were kept on a high-fat diet (20.1 kJ/g) for four weeks without or with addition of tiplaxtinin (PAI-039) at a dose of 2 mg/g food. At the time of sacrifice, body weights were significantly lower in the inhibitor-treated mice (p < 0.0005). The weights of the isolated subcutaneous and gonadal fat deposits were also significantly lower (both p < 0.0005), associated with adipocyte hypotrophy. Inhibitor-treated adipose tissues displayed similar blood vessel size, but a higher blood vessel density. Fasting glucose and insulin levels, as well as glucose-tolerance tests were not significantly affected by the inhibitor treatment, whereas plasma triglyceride levels were significantly reduced (p = 0.02) and LDL-cholesterol levels significantly enhanced (p = 0.0002). Insulin-tolerance tests revealed significantly lower glucose levels at the end of the test in the inhibitor treated mice (p = 0.03). Thus, in this model of diet-induced obesity in mice administration of tiplaxtinin resulted in impaired adipose tissue development.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Peso Corporal/efectos de los fármacos , Ácidos Indolacéticos/farmacología , Obesidad/prevención & control , Inhibidor 1 de Activador Plasminogénico/metabolismo , Tejido Adiposo/irrigación sanguínea , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Fármacos Antiobesidad/uso terapéutico , Glucemia/efectos de los fármacos , Grasas de la Dieta , Modelos Animales de Enfermedad , Ingestión de Energía , Fibrinólisis/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Obesidad/sangre , Obesidad/metabolismo , Obesidad/patología , Tamaño de los Órganos/efectos de los fármacos , Factores de Tiempo
19.
Biochim Biophys Acta ; 1760(12): 1837-44, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17011710

RESUMEN

The expression and potential functional role of aggrecan in adipogenesis and adipose tissue development was investigated in murine models of obesity. Aggrecan, as well as the two aggrecanases ADAMTS-4 and ADAMTS-5 (A Disintegrin And Metalloproteinase with Thrombospondin motif) mRNAs, are expressed in subcutaneous (SC) and gonadal (GON) adipose tissues of mice. Their presence was confirmed by western blotting using adipose tissue extracts. In mice with nutritionally induced obesity (high fat diet) as well as in lean controls, aggrecan mRNA expression was downregulated whereas ADAMTS-4 and ADAMTS-5 were upregulated with time. In mice with genetically determined obesity (ob/ob), ADAMTS-5 mRNA was upregulated in both SC and GON adipose tissues, as compared to wild-type (WT) mice (p<0.001). Enhanced aggrecanase expression levels in these tissues were associated with significantly elevated levels of G1-NITEGE, a degradation product of aggrecan. Thus, aggrecan levels were high at the early stages of adipose tissue development in mice, whereas its production decreased and its degradation increased during development of obesity. A functional role of aggrecan in promoting early stages of adipogenesis is supported by the findings that it stimulated the in vitro differentiation of 3T3-F442A preadipocytes and the de novo in vivo accumulation of fat in Matrigel plaques injected into WT mice. Proteoglycans in the extracellular matrix of adipose tissue, such as aggrecan, may contribute to the regulation of lipid uptake and obesity in mice.


Asunto(s)
Tejido Adiposo/crecimiento & desarrollo , Agrecanos/metabolismo , Diferenciación Celular , Endopeptidasas/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS4 , Proteína ADAMTS5 , Tejido Adiposo/citología , Agrecanos/genética , Animales , Secuencia de Bases , Cartilla de ADN , Endopeptidasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Procolágeno N-Endopeptidasa/genética , Procolágeno N-Endopeptidasa/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Diabetes ; 55(10): 2698-704, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17003333

RESUMEN

Placental growth factor (PlGF)-deficient (PlGF-/-) and wild-type mice were kept on a standard-fat or high-fat diet for 15 weeks. With the standard-fat diet, the body weights of PlGF-/- and wild-type mice were comparable, whereas the combined weight of subcutaneous and gonadal adipose tissues was lower in PlGF-/- mice (P = 0.02). With the high-fat diet, PlGF-/- mice had a lower body weight (P < 0.05) and less total subcutaneous plus gonadal adipose tissue (P < 0.0001). Blood vessel size was lower in gonadal adipose tissue of PlGF-/- mice with both the standard-fat and high-fat diet (P < 0.05). Blood vessel density, normalized to adipocyte number, was significantly lower in subcutaneous adipose tissue of PlGF-/- mice fed the high-fat diet (P < 0.01). De novo adipose tissue development in nude mice injected with 3T3-F442A preadipocytes was reduced (P < 0.005) by administration of a PlGF-neutralizing antibody. Bone marrow transplantation from wild-type or PlGF-/- mice to wild-type or PlGF-/- recipient mice revealed significantly lower blood vessel density in PlGF-/- recipient mice without an effect on adipose tissue growth. Thus, in murine models of diet-induced obesity, inactivation of PlGF impairs adipose tissue development, at least in part as a result of reduced angiogenesis.


Asunto(s)
Tejido Adiposo/crecimiento & desarrollo , Proteínas Gestacionales/deficiencia , Adipocitos/citología , Tejido Adiposo/irrigación sanguínea , Animales , Anticuerpos Monoclonales/farmacología , Trasplante de Médula Ósea/fisiología , Grasas de la Dieta/administración & dosificación , Masculino , Ratones , Ratones Desnudos , Factor de Crecimiento Placentario , Proteínas Gestacionales/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...