Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 378, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33432089

RESUMEN

Quantitative assessment of soft tissue elasticity is crucial to a broad range of applications, such as biomechanical modeling, physiological monitoring, and tissue diseases diagnosing. However, the modulus measurement of soft tissues, particularly in vivo, has proved challenging since the instrument has to reach the site of soft tissue and be able to measure in a very short time. Here, we present a simple method to measure the elastic modulus of soft tissues on site by exploiting buckling of a long slender bar to quantify the applied force and a spherical indentation to extract the elastic modulus. The method is realized by developing a portable pen-sized instrument (EPen: Elastic modulus pen). The measurement accuracies are verified by independent modulus measures using commercial nanoindenter. Quantitative measurements of the elastic modulus of mouse pancreas, healthy and cancerous, surgically exposed but attached to the body further confirm the potential clinical utility of the EPen.


Asunto(s)
Estructuras Animales/fisiología , Fenómenos Biomecánicos/fisiología , Elasticidad/fisiología , Tecnología de Fibra Óptica/instrumentación , Animales , Biofisica/instrumentación , Módulo de Elasticidad , Femenino , Tecnología de Fibra Óptica/métodos , Ensayo de Materiales , Ratones , Ratones Transgénicos , Microtecnología/instrumentación , Aplicaciones Móviles , Tono Muscular/fisiología , Fenómenos Fisiológicos Musculoesqueléticos , Agujas , Estrés Mecánico
2.
Proc Natl Acad Sci U S A ; 116(29): 14724-14733, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31266893

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is notorious for its poor survival and resistance to conventional therapies. PI3K signaling is implicated in both disease initiation and progression, and specific inhibitors of selected PI3K p110 isoforms for managing solid tumors are emerging. We demonstrate that increased activation of PI3K signals cooperates with oncogenic Kras to promote aggressive PDAC in vivo. The p110γ isoform is overexpressed in tumor tissue and promotes carcinogenesis via canonical AKT signaling. Its selective blockade sensitizes tumor cells to gemcitabine in vitro, and genetic ablation of p110γ protects against Kras-induced tumorigenesis. Diet/obesity was identified as a crucial means of p110 subunit up-regulation, and in the setting of a high-fat diet, p110γ ablation failed to protect against tumor development, showing increased activation of pAKT and hepatic damage. These observations suggest that a careful and judicious approach should be considered when targeting p110γ for therapy, particularly in obese patients.


Asunto(s)
Carcinogénesis/genética , Carcinoma Ductal Pancreático/genética , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/complicaciones , Neoplasias Pancreáticas/genética , Animales , Antimetabolitos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/uso terapéutico , Carcinogénesis/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Ácidos Grasos Omega-6/efectos adversos , Femenino , Glucosa/metabolismo , Humanos , Metabolismo de los Lípidos , Hígado/patología , Masculino , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Páncreas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Regulación hacia Arriba , Gemcitabina
3.
Mol Cancer Ther ; 18(3): 613-620, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30587556

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) remains remarkably lethal with a 5-year survival rate of 8%. This is mainly attributed to the late stage of presentation, as well as widespread resistance to conventional therapy. In addition, PDAC tumors are largely nonimmunogenic, and most patients have displayed incomplete responses to cancer immunotherapies. Our group has previously identified TGFß as a crucial repressor of antitumor immune function in PDAC, particularly with respect to cytotoxic T lymphocytes. However, pharmacologic inhibition of TGFß signaling has had limited efficacy in clinical trials, failing to promote a significant antitumor immune response. Hence, in this work, we extend our analysis to identify and circumvent the mechanisms of resistance to TGFß signal inhibition in PDAC. Consistent with our previous observations, adoptive transfer of TGFß-insensitive CD8+ T cells led to the near complete regression of neoplastic disease in vivo However, we demonstrate that this cannot be recapitulated via global reduction in TGFß signaling, through either genetic ablation or pharmacologic inhibition of TGFBR1. In fact, tumors with TGFß signal inhibition displayed increased PD-L1 expression and had no observable change in antitumor immunity. Using genetic models of advanced PDAC, we then determined that concomitant inhibition of both TGFß and PD-L1 receptors led to a reduction in the neoplastic phenotype, improving survival and reducing disease-associated morbidity in vivo Combined, these data strongly suggest that TGFß and PD-L1 pathway inhibitors may synergize in PDAC, and this approach warrants clinical consideration.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antígeno B7-H1/genética , Carcinoma Ductal Pancreático/tratamiento farmacológico , Factor de Crecimiento Transformador beta/genética , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Antígeno B7-H1/antagonistas & inhibidores , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoterapia , Ratones , Ratones Transgénicos , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Microambiente Tumoral/efectos de los fármacos
4.
Nutrients ; 10(9)2018 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-30213082

RESUMEN

Pancreatic cancer remains a daunting foe despite a vast number of accumulating molecular analyses regarding the mutation and expression status of a variety of genes. Indeed, most pancreatic cancer cases uniformly present with a mutation in the KRAS allele leading to enhanced RAS activation. Yet our understanding of the many epigenetic/environmental factors contributing to disease incidence and progression is waning. Epidemiologic data suggest that diet may be a key factor in pancreatic cancer development and potentially a means of chemoprevention at earlier stages. While diets high in ω3 fatty acids are typically associated with tumor suppression, diets high in ω6 fatty acids have been linked to increased tumor development. Thus, to better understand the contribution of these polyunsaturated fatty acids to pancreatic carcinogenesis, we modeled early stage disease by targeting mutant KRAS to the exocrine pancreas and administered diets rich in these fatty acids to assess tumor formation and altered cell-signaling pathways. We discovered that, consistent with previous reports, the ω3-enriched diet led to reduced lesion penetrance via repression of proliferation associated with reduced phosphorylated AKT (pAKT), whereas the ω6-enriched diet accelerated tumor formation. These data provide a plausible mechanism underlying previously observed effects of fatty acids and suggest that administration of ω3 fatty acids can reduce the pro-survival, pro-growth functions of pAKT. Indeed, counseling subjects at risk to increase their intake of foods containing higher amounts of ω3 fatty acids could aid in the prevention of pancreatic cancer.


Asunto(s)
Anticarcinógenos/administración & dosificación , Transformación Celular Neoplásica/metabolismo , Dieta , Ácidos Grasos Omega-3/administración & dosificación , Neoplasias Experimentales/prevención & control , Conductos Pancreáticos/enzimología , Neoplasias Pancreáticas/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Apoptosis , Línea Celular , Proliferación Celular , Supervivencia Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Dieta/efectos adversos , Regulación hacia Abajo , Humanos , Ratones Transgénicos , Mutación , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fosforilación , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
5.
J Immunol ; 201(8): 2414-2426, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30201810

RESUMEN

Ischemic tissue damage activates hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM)-generating myeloid cells, and persistent HSPC activity may drive chronic inflammation and impair tissue recovery. Although increased reactive oxygen species in the BM regulate HSPC functions, their roles in myelopoiesis of activated HSPCs and subsequent tissue recovery during ischemic damage are not well understood. In this paper, we report that deletion of Nox2 NADPH oxidase in mice results in persistent elevations in BM HSPC activity and levels of inflammatory monocytes/macrophages in BM and ischemic tissue in a model of hindlimb ischemia. Ischemic tissue damage induces oxidants in BM such as elevations of hydrogen peroxide and oxidized phospholipids, which activate redox-sensitive Lyn kinase in a Nox2-dependent manner. Moreover, during tissue recovery after ischemic injury, this Nox2-ROS-Lyn kinase axis is induced by Nox2 in neutrophils that home to the BM, which inhibits HSPC activity and inflammatory monocyte generation and promotes tissue regeneration after ischemic damage. Thus, oxidant signaling in the BM mediated by Nox2 in neutrophils regulates myelopoiesis of HSPCs to promote regeneration of damaged tissue.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Miembro Posterior/patología , Isquemia/inmunología , NADPH Oxidasa 2/metabolismo , Neutrófilos/fisiología , Animales , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mielopoyesis , NADPH Oxidasa 2/genética , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Regeneración , Transducción de Señal , Familia-src Quinasas/metabolismo
6.
Sci Rep ; 8(1): 12548, 2018 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-30135483

RESUMEN

Although survival has improved in recent years, the prognosis of patients with advanced pancreatic ductal adenocarcinoma (PDAC) remains poor. Despite substantial differences in anatomy, physiology, genetics, and metabolism, the overwhelming majority of preclinical testing relies on transgenic mice. Hence, while mice have allowed for tremendous advances in cancer biology, they have been a poor predictor of drug performance/toxicity in the clinic. Given the greater similarity of sus scrofa pigs to humans, we engineered transgenic sus scrofa expressing a LSL-KRASG12D-TP53R167H cassette. By applying Adeno-Cre to pancreatic duct cells in vitro, cells self-immortalized and established tumors in immunocompromised mice. When Adeno-Cre was administered to the main pancreatic duct in vivo, pigs developed extensive PDAC at the injection site hallmarked by excessive proliferation and desmoplastic stroma. This serves as the first large animal model of pancreatic carcinogenesis, and may allow for insight into new avenues of translational research not before possible in rodents.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Sus scrofa/genética , Proteína p53 Supresora de Tumor/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Animales Modificados Genéticamente , Carcinoma Ductal Pancreático/patología , Femenino , Humanos , Integrasas , Ratones SCID , Mutación , Neoplasias Experimentales , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/patología , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncotarget ; 8(54): 92667-92681, 2017 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-29190947

RESUMEN

Peroxiredoxin-1 (Prdx1), a member of the thioredoxin (Txn) system, is overexpressed and correlates with poor prognosis in pancreatic cancer patients and can suppress Kras signaling through redox-mediated inhibition of ERK and AKT in lung and breast cancer. Its redox function is maintained by Txn and sulfiredoxin (Srxn), and its tumor promoting functions are activated by post-translational modification. We studied the role of the Txn system in pancreatic neoplasia and cancer by determining how it regulates the phosphorylation of Kras effectors and by determining its association with patient survival. We found that elevated Prdx1 nuclear localization significantly correlated with better patient survival. Our data also demonstrate that the expression of the Txn system is dysregulated, with elevated Prdx1 expression and significantly decreased Txn and Srxn expression in pancreatic lesions of targeted mutant Kras mouse models. This correlated with distinct differences in the interconversion of Prdx1 oligomers that affect its ability to regulate ERK and AKT phosphorylation. Our data also suggest that Prdx1 post-translational modification and oligomerization suppress Prdx1 mediated redox regulation of ERK phosphorylation. We observed distinct differences in Txn expression and in the ability of pTyr-Prdx1 to bind to pERK in a PanIN model of pancreatic neoplasia as compared to an IPMN model, indicating a distinct difference in the function of post-translationally modified Prdx1 in cells with less Txn expression. Modified Txn system function and post-translational regulation may therefore play a significant role in pancreatic tumorigenesis by altering Kras effector phosphorylation and inhibiting the tumor suppressive redox functions of Prdx1.

8.
Oncotarget ; 8(3): 3826-3839, 2017 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-27270652

RESUMEN

TGFß has both tumor suppressive and tumor promoting effects in colon cancer. Also, TGFß can affect the extent and composition of inflammatory cells present in tumors, contextually promoting and inhibiting inflammation. While colon tumors display intratumoral inflammation, the contributions of TGFß to this process are poorly understood. In human patients, we found that epithelial loss of TGFß signaling was associated with increased inflammatory burden; yet overexpression of TGFß was also associated with increased inflammation. These findings were recapitulated in mutant APC models of murine tumorigenesis, where epithelial truncation of TGFBR2 led to lethal inflammatory disease and invasive colon cancer, mediated by IL8 and TGFß1. Interestingly, mutant APC mice with global suppression of TGFß signals displayed an intermediate phenotype, presenting with an overall increase in IL8-mediated inflammation and accelerated tumor formation, yet with a longer latency to the onset of disease observed in mice with epithelial TGFBR-deficiency. These results suggest that the loss of TGFß signaling, particularly in colon epithelial cells, elicits a strong inflammatory response and promotes tumor progression. This implies that treating colon cancer patients with TGFß inhibitors may result in a worse outcome by enhancing inflammatory responses.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Neoplasias del Colon/patología , Citocinas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Neoplasias Experimentales , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Proteína Smad4/metabolismo
9.
Sci Rep ; 5: 14780, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26437801

RESUMEN

Copper (Cu), an essential micronutrient, plays a fundamental role in inflammation and angiogenesis; however, its precise mechanism remains undefined. Here we uncover a novel role of Cu transport protein Antioxidant-1 (Atox1), which is originally appreciated as a Cu chaperone and recently discovered as a Cu-dependent transcription factor, in inflammatory neovascularization. Atox1 expression is upregulated in patients and mice with critical limb ischemia. Atox1-deficient mice show impaired limb perfusion recovery with reduced arteriogenesis, angiogenesis, and recruitment of inflammatory cells. In vivo intravital microscopy, bone marrow reconstitution, and Atox1 gene transfer in Atox1(-/-) mice show that Atox1 in endothelial cells (ECs) is essential for neovascularization and recruitment of inflammatory cells which release VEGF and TNFα. Mechanistically, Atox1-depleted ECs demonstrate that Cu chaperone function of Atox1 mediated through Cu transporter ATP7A is required for VEGF-induced angiogenesis via activation of Cu enzyme lysyl oxidase. Moreover, Atox1 functions as a Cu-dependent transcription factor for NADPH oxidase organizer p47phox, thereby increasing ROS-NFκB-VCAM-1/ICAM-1 expression and monocyte adhesion in ECs inflamed with TNFα in an ATP7A-independent manner. These findings demonstrate a novel linkage between Atox1 and NADPH oxidase involved in inflammatory neovascularization and suggest Atox1 as a potential therapeutic target for treatment of ischemic disease.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Transporte de Catión/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Isquemia/genética , Metalochaperonas/genética , NADPH Oxidasas/genética , Neovascularización Patológica/genética , Proteína-Lisina 6-Oxidasa/genética , Adenosina Trifosfatasas/metabolismo , Animales , Proteínas de Transporte de Catión/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Proteínas Transportadoras de Cobre , ATPasas Transportadoras de Cobre , Regulación de la Expresión Génica , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Isquemia/metabolismo , Isquemia/patología , Pierna/irrigación sanguínea , Pierna/patología , Metalochaperonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares , Monocitos/metabolismo , Monocitos/patología , NADPH Oxidasas/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Proteína-Lisina 6-Oxidasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Diabetes ; 62(11): 3839-50, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23884884

RESUMEN

Oxidative stress and endothelial dysfunction contribute to vascular complication in diabetes. Extracellular superoxide dismutase (SOD3) is one of the key antioxidant enzymes that obtains copper via copper transporter ATP7A. SOD3 is secreted from vascular smooth muscles cells (VSMCs) and anchors at the endothelial surface. The role of SOD3 and ATP7A in endothelial dysfunction in type 1 diabetes mellitus (T1DM) is entirely unknown. Here we show that the specific activity of SOD3, but not SOD1, is decreased, which is associated with increased O2(•-) production in aortas of streptozotocin-induced and genetically induced Ins2(Akita) T1DM mice. Exogenous copper partially rescued SOD3 activity in isolated T1DM vessels. Functionally, acetylcholine-induced, endothelium-dependent relaxation is impaired in T1DM mesenteric arteries, which is rescued by SOD mimetic tempol or gene transfer of SOD3. Mechanistically, ATP7A expression in T1DM vessels is dramatically decreased whereas other copper transport proteins are not altered. T1DM-induced endothelial dysfunction and decrease of SOD3 activity are rescued in transgenic mice overexpressing ATP7A. Furthermore, SOD3-deficient T1DM mice or ATP7A mutant T1DM mice augment endothelial dysfunction and vascular O2(•-) production versus T1DM mice. These effects are in part due to hypoinsulinemia in T1DM mice, since insulin treatment, but not high glucose, increases ATP7A expression in VSMCs and restores SOD3 activity in the organoid culture of T1DM vessels. In summary, a decrease in ATP7A protein expression contributes to impaired SOD3 activity, resulting in O2(•-) overproduction and endothelial dysfunction in blood vessels of T1DM. Thus, restoring copper transporter function is an essential therapeutic approach for oxidant stress-dependent vascular and metabolic diseases.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Proteínas de Transporte de Catión/fisiología , Cobre/metabolismo , Superóxido Dismutasa/metabolismo , Adenosina Trifosfatasas/genética , Animales , Aorta/metabolismo , Proteínas de Transporte de Catión/genética , Células Cultivadas , Cobre/farmacología , ATPasas Transportadoras de Cobre , Óxidos N-Cíclicos/farmacología , Diabetes Mellitus Experimental/fisiopatología , Insulina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Ratas , Marcadores de Spin , Superóxido Dismutasa/genética , Superóxidos/metabolismo , Transfección
11.
Am J Physiol Cell Physiol ; 305(6): C591-600, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23657573

RESUMEN

Platelet-derived growth factor (PDGF) stimulates vascular smooth muscle cell (VSMC) migration and neointimal formation in response to injury. We previously identified IQ-domain GTPase-activating protein 1 (IQGAP1) as a novel VEGF receptor 2 binding scaffold protein involved in endothelial migration. However, its role in VSMC migration and neointimal formation in vivo is unknown. Here we show that PDGF stimulation rapidly promotes IQGAP1 association with PDGF receptor-ß (PDGFR) as well as IQGAP1 tyrosine phosphorylation in cultured VSMC. Overexpression or knockdown of IQGAP1 enhances or inhibits PDGFR autophosphorylation (p-PDGFR), respectively. Immunofluorescence and cell fractionation analysis reveals that PDGF-induced p-PDGFR localized in focal adhesions (FAs), but not caveolae/lipid rafts, is inhibited by IQGAP1 knockdown with siRNA. PDGF stimulation promotes IQGAP1 association with PDGFR/FA signaling protein complex. Functionally, IQGAP1 siRNA inhibits PDGF-induced FA formation as well as VSMC migration induced by PDGF. In vivo, IQGAP1 expression is markedly increased at neointimal VSMC in wire-injured femoral arteries. Mice lacking IQGAP1 exhibit impaired neointimal formation in response to vascular injury. In summary, IQGAP1, through interaction with PDGFR and FA signaling proteins, promotes activation of PDGFR in FAs as well as FA formation, which may contribute to VSMC migration and neointimal formation after injury. Our findings provide insight into IQGAP1 as a potential therapeutic target for vascular migration-related diseases.


Asunto(s)
Movimiento Celular/fisiología , Adhesiones Focales/fisiología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Neointima/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Células Cultivadas , Arteria Femoral/metabolismo , Arteria Femoral/fisiología , Adhesiones Focales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Fosforilación/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Ratas , Ratas Sprague-Dawley , Tirosina/metabolismo , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
12.
PLoS One ; 8(3): e57618, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23472092

RESUMEN

BACKGROUND: Reactive oxygen species (ROS) play an important role in angiogenesis in endothelial cells (ECs) in vitro and neovascularization in vivo. However, little is known about the role of endogenous vascular hydrogen peroxide (H2O2) in postnatal neovascularization. METHODOLOGY/PRINCIPAL FINDINGS: We used Tie2-driven endothelial specific catalase transgenic mice (Cat-Tg mice) and hindlimb ischemia model to address the role of endogenous H2O2 in ECs in post-ischemic neovascularization in vivo. Here we show that Cat-Tg mice exhibit significant reduction in intracellular H2O2 in ECs, blood flow recovery, capillary formation, collateral remodeling with larger extent of tissue damage after hindlimb ischemia, as compared to wild-type (WT) littermates. In the early stage of ischemia-induced angiogenesis, Cat-Tg mice show a morphologically disorganized microvasculature. Vascular sprouting and tube elongation are significantly impaired in isolated aorta from Cat-Tg mice. Furthermore, Cat-Tg mice show a decrease in myeloid cell recruitment after hindlimb ischemia. Mechanistically, Cat-Tg mice show significant decrease in eNOS phosphorylation at Ser1177 as well as expression of redox-sensitive vascular cell adhesion molecule-1 (VCAM-1) and monocyte chemotactic protein-1 (MCP-1) in ischemic muscles, which is required for inflammatory cell recruitment to the ischemic tissues. We also observed impaired endothelium-dependent relaxation in resistant vessels from Cat-Tg mice. CONCLUSIONS/SIGNIFICANCE: Endogenous ECs-derived H2O2 plays a critical role in reparative neovascularization in response to ischemia by upregulating adhesion molecules and activating eNOS in ECs. Redox-regulation in ECs is a potential therapeutic strategy for angiogenesis-dependent cardiovascular diseases.


Asunto(s)
Células Endoteliales/metabolismo , Peróxido de Hidrógeno/metabolismo , Isquemia/patología , Neovascularización Fisiológica , Animales , Catalasa/metabolismo , Quimiocina CCL2/metabolismo , Ratones , Ratones Transgénicos , Microcirculación , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Oxidación-Reducción , Fosforilación , Especies Reactivas de Oxígeno , Receptor TIE-2/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 33(4): 805-13, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23349186

RESUMEN

OBJECTIVE: Vascular smooth muscle cell (VSMC) migration is critically important for neointimal formation after vascular injury and atherosclerosis lesion formation. Copper (Cu) chelator inhibits neointimal formation, and we previously demonstrated that Cu transport protein antioxidant-1 (Atox1) is involved in Cu-induced cell growth. However, role of Atox1 in VSMC migration and neointimal formation after vascular injury is unknown. APPROACH AND RESULTS: Here, we show that Atox1 expression is upregulated in injured vessel, and it is colocalized with the Cu transporter ATP7A, one of the downstream targets of Atox1, mainly in neointimal VSMCs at day 14 after wire injury. Atox1(-/-) mice show inhibition of neointimal formation and extracellular matrix expansion, which is associated with a decreased VSMCs accumulation within neointima and lysyl oxidase activity. Mechanistically, in cultured VSMC, Atox1 depletion with siRNA inhibits platelet-derived growth factor-induced Cu-dependent VSMC migration by preventing translocation of ATP7A and small G protein Rac1 to the leading edge, as well as Cu- and Rac1-dependent lamellipodia formation. Furthermore, Atox1(-/-) mice show decreased perivascular macrophage infiltration in wire-injured vessels, as well as thioglycollate-induced peritoneal macrophage recruitment. CONCLUSIONS: Atox1 is involved in neointimal formation after vascular injury through promoting VSMC migration and inflammatory cell recruitment in injured vessels. Thus, Atox1 is a potential therapeutic target for VSMC migration and inflammation-related vascular diseases.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Chaperonas Moleculares/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima , Lesiones del Sistema Vascular/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Movimiento Celular , Células Cultivadas , Proteínas Transportadoras de Cobre , ATPasas Transportadoras de Cobre , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Arteria Femoral/patología , Humanos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Noqueados , Chaperonas Moleculares/genética , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/patología , Neuropéptidos/metabolismo , Peritonitis/inducido químicamente , Peritonitis/inmunología , Peritonitis/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transporte de Proteínas , Proteína-Lisina 6-Oxidasa/metabolismo , Seudópodos/metabolismo , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Tioglicolatos , Factores de Tiempo , Transfección , Regulación hacia Arriba , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/inmunología , Lesiones del Sistema Vascular/patología , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
14.
Hypertension ; 60(2): 476-86, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22753205

RESUMEN

Extracellular superoxide dismutase (SOD3) is a secretory copper enzyme involved in protecting angiotensin II (Ang II)-induced hypertension. We found previously that Ang II upregulates SOD3 expression and activity as a counterregulatory mechanism; however, underlying mechanisms are unclear. Antioxidant 1 (Atox1) is shown to act as a copper-dependent transcription factor, as well as a copper chaperone, for SOD3 in vitro, but its role in Ang II-induced hypertension in vivo is unknown. Here we show that Ang II infusion increases Atox1 expression, as well as SOD3 expression and activity, in aortas of wild-type mice, which are inhibited in mice lacking Atox1. Accordingly, Ang II increases vascular superoxide production, reduces endothelium-dependent vasodilation, and increases vasoconstriction in mesenteric arteries to a greater extent in Atox1(-/-) than in wild-type mice. This contributes to augmented hypertensive response to Ang II in Atox1(-/-) mice. In cultured vascular smooth muscle cells, Ang II promotes translocation of Atox1 to the nucleus, thereby increasing SOD3 transcription by binding to Atox1-responsive element in the SOD3 promoter. Furthermore, Ang II increases Atox1 binding to the copper exporter ATP7A, which obtains copper from Atox1, as well as translocation of ATP7A to plasma membranes, where it colocalizes with SOD3. As its consequence, Ang II decreases vascular copper levels, which is inhibited in Atox1(-/-) mice. In summary, Atox1 functions to prevent Ang II-induced endothelial dysfunction and hypercontraction in resistant vessels, as well as hypertension, in vivo by reducing extracellular superoxide levels via increasing vascular SOD3 expression and activity.


Asunto(s)
Angiotensina II/efectos adversos , Proteínas de Transporte de Catión/metabolismo , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Chaperonas Moleculares/metabolismo , Superóxido Dismutasa/metabolismo , Adenosina Trifosfatasas/metabolismo , Angiotensina II/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Cobre/metabolismo , Proteínas Transportadoras de Cobre , ATPasas Transportadoras de Cobre , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Oxígeno/metabolismo
15.
Stem Cells ; 30(5): 923-34, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22290850

RESUMEN

Bone marrow (BM) microenvironment, which is regulated by hypoxia and proteolytic enzymes, is crucial for stem/progenitor cell function and mobilization involved in postnatal neovascularization. We demonstrated that NADPH oxidase 2 (Nox2)-derived reactive oxygen species (ROS) are involved in postischemic mobilization of BM cells and revascularization. However, role of Nox2 in regulating BM microenvironment in response to ischemic injury remains unknown. Here, we show that hindlimb ischemia of mice increases ROS production in both the endosteal and central region of BM tissue in situ, which is almost completely abolished in Nox2 knockout (KO) mice. This Nox2-dependent ROS production is mainly derived from Gr-1(+) myeloid cells in BM. In vivo injection of hypoxyprobe reveals that endosteum at the BM is hypoxic with high expression of hypoxia-inducible factor-1α in basal state. Following hindlimb ischemia, hypoxic areas and HIF-1α expression are expanded throughout the BM, which is inhibited in Nox2 KO mice. This ischemia-induced alteration of Nox2-dependent BM microenvironment is associated with an increase in vascular endothelial growth factor expression and Akt phosphorylation in BM tissue, thereby promoting Lin(-) progenitor cell survival and expansion, leading to their mobilization from BM. Furthermore, hindlimb ischemia increases proteolytic enzymes membrane type 1-matrix metalloproteinase (MMP) expression and MMP-9 activity in BM, which is inhibited in Nox2 KO mice. In summary, Nox2-dependent increase in ROS plays a critical role in regulating hypoxia expansion and proteolytic activities in BM microenvironment in response to tissue ischemia. This in turn promotes progenitor cell expansion and reparative mobilization from BM, leading to postischemic neovascularization and tissue repair.


Asunto(s)
Médula Ósea/enzimología , Miembro Posterior/irrigación sanguínea , Isquemia/enzimología , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasas/metabolismo , Regeneración , Nicho de Células Madre , Células Madre/enzimología , Animales , Médula Ósea/patología , Miembro Posterior/metabolismo , Miembro Posterior/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isquemia/genética , Isquemia/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , NADPH Oxidasa 2 , NADPH Oxidasas/genética , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células Madre/patología
16.
Am J Physiol Heart Circ Physiol ; 302(3): H724-32, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22101521

RESUMEN

p66Shc, a longevity adaptor protein, is demonstrated as a key regulator of reactive oxygen species (ROS) metabolism involved in aging and cardiovascular diseases. Vascular endothelial growth factor (VEGF) stimulates endothelial cell (EC) migration and proliferation primarily through the VEGF receptor-2 (VEGFR2). We have shown that ROS derived from Rac1-dependent NADPH oxidase are involved in VEGFR2 autophosphorylation and angiogenic-related responses in ECs. However, a role of p66Shc in VEGF signaling and physiological responses in ECs is unknown. Here we show that VEGF promotes p66Shc phosphorylation at Ser36 through the JNK/ERK or PKC pathway as well as Rac1 binding to a nonphosphorylated form of p66Shc in ECs. Depletion of endogenous p66Shc with short interfering RNA inhibits VEGF-induced Rac1 activity and ROS production. Fractionation of caveolin-enriched lipid raft demonstrates that p66Shc plays a critical role in VEGFR2 phosphorylation in caveolae/lipid rafts as well as downstream p38MAP kinase activation. This in turn stimulates VEGF-induced EC migration, proliferation, and capillary-like tube formation. These studies uncover a novel role of p66Shc as a positive regulator for ROS-dependent VEGFR2 signaling linked to angiogenesis in ECs and suggest p66Shc as a potential therapeutic target for various angiogenesis-dependent diseases.


Asunto(s)
Células Endoteliales/enzimología , Sistema de Señalización de MAP Quinasas/fisiología , Neovascularización Fisiológica/fisiología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Caveolas/enzimología , Células Endoteliales/citología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Microdominios de Membrana/enzimología , Fosforilación/efectos de los fármacos , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteína de Unión al GTP rac1/metabolismo
17.
Free Radic Res ; 45(10): 1124-35, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21740309

RESUMEN

Reactive oxygen species (ROS) are important mediators for VEGF receptor 2 (VEGFR2) signalling involved in angiogenesis. The initial product of Cys oxidation, cysteine sulfenic acid (Cys-OH), is a key intermediate in redox signal transduction; however, its role in VEGF signalling is unknown. We have previously demonstrated IQGAP1 as a VEGFR2 binding scaffold protein involved in ROS-dependent EC migration and post-ischemic angiogenesis. Using a biotin-labelled Cys-OH trapping reagent, we show that VEGF increases protein-Cys-OH formation at the lamellipodial leading edge where it co-localizes with NADPH oxidase and IQGAP1 in migrating ECs, which is prevented by IQGAP1 siRNA or trapping of Cys-OH with dimedone. VEGF increases IQGAP1-Cys-OH formation, which is prevented by N-acetyl cysteine or dimedone, which inhibits VEGF-induced EC migration and capillary network formation. In vivo, hindlimb ischemia in mice increases Cys-OH formation in small vessels and IQGAP1 in ischemic tissues. In summary, VEGF stimulates localized formation of Cys-OH-IQGAP1 at the leading edge, thereby promoting directional EC migration, which may contribute to post-natal angiogenesis in vivo. Thus, targeting Cys-oxidized proteins at specific compartments may be the potential therapeutic strategy for various angiogenesis-dependent diseases.


Asunto(s)
Movimiento Celular/fisiología , Cisteína/análogos & derivados , Células Endoteliales/citología , Células Endoteliales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Actinas/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Cisteína/biosíntesis , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , NADPH Oxidasas/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Ácidos Sulfénicos , Transfección , Factor A de Crecimiento Endotelial Vascular/farmacología , Proteínas Activadoras de ras GTPasa/biosíntesis , Proteínas Activadoras de ras GTPasa/genética , Proteínas Activadoras de ras GTPasa/metabolismo
18.
PLoS One ; 5(10): e13440, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20976168

RESUMEN

BACKGROUND: Neovascularization is an important repair mechanism in response to ischemic injury and is dependent on inflammation, angiogenesis and reactive oxygen species (ROS). IQGAP1, an actin-binding scaffold protein, is a key regulator for actin cytoskeleton and motility. We previously demonstrated that IQGAP1 mediates vascular endothelial growth factor (VEGF)-induced ROS production and migration of cultured endothelial cells (ECs); however, its role in post-ischemic neovascularization is unknown. METHODOLOGY/PRINCIPAL FINDINGS: Ischemia was induced by left femoral artery ligation, which resulted in increased IQGAP1 expression in Mac3(+) macrophages and CD31(+) capillary-like ECs in ischemic legs. Mice lacking IQGAP1 exhibited a significant reduction in the post-ischemic neovascularization as evaluated by laser Doppler blood flow, capillary density and α-actin positive arterioles. Furthermore, IQGAP1(-/-) mice showed a decrease in macrophage infiltration and ROS production in ischemic muscles, leading to impaired muscle regeneration and increased necrosis and fibrosis. The numbers of bone marrow (BM)-derived cells in the peripheral blood were not affected in these knockout mice. BM transplantation revealed that IQGAP1 expressed in both BM-derived cells and tissue resident cells, such as ECs, is required for post-ischemic neovascularization. Moreover, thioglycollate-induced peritoneal macrophage recruitment and ROS production were inhibited in IQGAP1(-/-) mice. In vitro, IQGAP1(-/-) BM-derived macrophages showed inhibition of migration and adhesion capacity, which may explain the defective macrophage recruitment into the ischemic tissue in IQGAP1(-/-) mice. CONCLUSIONS/SIGNIFICANCE: IQGAP1 plays a key role in post-ischemic neovascularization by regulating, not only, ECs-mediated angiogenesis but also macrophage infiltration as well as ROS production. Thus, IQGAP1 is a potential therapeutic target for inflammation- and angiogenesis-dependent ischemic cardiovascular diseases.


Asunto(s)
Macrófagos/patología , Neovascularización Patológica , Proteínas Activadoras de ras GTPasa/fisiología , Animales , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Activadoras de ras GTPasa/genética
19.
Circ Res ; 107(6): 787-99, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20671235

RESUMEN

RATIONALE: Copper, an essential nutrient, has been implicated in vascular remodeling and atherosclerosis with unknown mechanism. Bioavailability of intracellular copper is regulated not only by the copper importer CTR1 (copper transporter 1) but also by the copper exporter ATP7A (Menkes ATPase), whose function is achieved through copper-dependent translocation from trans-Golgi network (TGN). Platelet-derived growth factor (PDGF) promotes vascular smooth muscle cell (VSMC) migration, a key component of neointimal formation. OBJECTIVE: To determine the role of copper transporter ATP7A in PDGF-induced VSMC migration. METHODS AND RESULTS: Depletion of ATP7A inhibited VSMC migration in response to PDGF or wound scratch in a CTR1/copper-dependent manner. PDGF stimulation promoted ATP7A translocation from the TGN to lipid rafts, which localized at the leading edge, where it colocalized with PDGF receptor and Rac1, in migrating VSMCs. Mechanistically, ATP7A small interfering RNA or CTR small interfering RNA prevented PDGF-induced Rac1 translocation to the leading edge, thereby inhibiting lamellipodia formation. In addition, ATP7A depletion prevented a PDGF-induced decrease in copper level and secretory copper enzyme precursor prolysyl oxidase (Pro-LOX) in lipid raft fraction, as well as PDGF-induced increase in LOX activity. In vivo, ATP7A expression was markedly increased and copper accumulation was observed by synchrotron-based x-ray fluorescence microscopy at neointimal VSMCs in wire injury model. CONCLUSIONS: These findings suggest that ATP7A plays an important role in copper-dependent PDGF-stimulated VSMC migration via recruiting Rac1 to lipid rafts at the leading edge, as well as regulating LOX activity. This may contribute to neointimal formation after vascular injury. Our findings provide insight into ATP7A as a novel therapeutic target for vascular remodeling and atherosclerosis.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Proteínas de Transporte de Catión/fisiología , Movimiento Celular/fisiología , Cobre/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Aterosclerosis/enzimología , Aterosclerosis/patología , Células Cultivadas , ATPasas Transportadoras de Cobre , Humanos , Masculino , Microdominios de Membrana/enzimología , Microdominios de Membrana/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/fisiología , Ratas , Ratas Sprague-Dawley
20.
PLoS One ; 5(4): e10189, 2010 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-20422004

RESUMEN

Reactive oxygen species (ROS), in particular, H(2)O(2), is essential for full activation of VEGF receptor2 (VEGFR2) signaling involved in endothelial cell (EC) proliferation and migration. Extracellular superoxide dismutase (ecSOD) is a major secreted extracellular enzyme that catalyzes the dismutation of superoxide to H(2)O(2), and anchors to EC surface through heparin-binding domain (HBD). Mice lacking ecSOD show impaired postnatal angiogenesis. However, it is unknown whether ecSOD-derived H(2)O(2) regulates VEGF signaling. Here we show that gene transfer of ecSOD, but not ecSOD lacking HBD (ecSOD-DeltaHBD), increases H(2)O(2) levels in adductor muscle of mice, and promotes angiogenesis after hindlimb ischemia. Mice lacking ecSOD show reduction of H(2)O(2) in non-ischemic and ischemic limbs. In vitro, overexpression of ecSOD, but not ecSOD-DeltaHBD, in cultured medium in ECs enhances VEGF-induced tyrosine phosphorylation of VEGFR2 (VEGFR2-pY), which is prevented by short-term pretreatment with catalase that scavenges extracellular H(2)O(2). Either exogenous H(2)O(2) (<500 microM), which is diffusible, or nitric oxide donor has no effect on VEGF-induced VEGFR2-pY. These suggest that ecSOD binding to ECs via HBD is required for localized generation of extracellular H(2)O(2) to regulate VEGFR2-pY. Mechanistically, VEGF-induced VEGFR2-pY in caveolae/lipid rafts, but non-lipid rafts, is enhanced by ecSOD, which localizes at lipid rafts via HBD. One of the targets of ROS is protein tyrosine phosphatases (PTPs). ecSOD induces oxidation and inactivation of both PTP1B and DEP1, which negatively regulates VEGFR2-pY, in caveolae/lipid rafts, but not non-lipid rafts. Disruption of caveolae/lipid rafts, or PTPs inhibitor orthovanadate, or siRNAs for PTP1B and DEP1 enhances VEGF-induced VEGFR2-pY, which prevents ecSOD-induced effect. Functionally, ecSOD promotes VEGF-stimulated EC migration and proliferation. In summary, extracellular H(2)O(2) generated by ecSOD localized at caveolae/lipid rafts via HBD promotes VEGFR2 signaling via oxidative inactivation of PTPs in these microdomains. Thus, ecSOD is a potential therapeutic target for angiogenesis-dependent cardiovascular diseases.


Asunto(s)
Caveolas/metabolismo , Peróxido de Hidrógeno/metabolismo , Isquemia/patología , Neovascularización Fisiológica , Superóxido Dismutasa/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Células Endoteliales/citología , Microdominios de Membrana/metabolismo , Ratones , Oxidación-Reducción , Fosforilación , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA