Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 578(7794): 290-295, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32025034

RESUMEN

Shear stress on arteries produced by blood flow is important for vascular development and homeostasis but can also initiate atherosclerosis1. Endothelial cells that line the vasculature use molecular mechanosensors to directly detect shear stress profiles that will ultimately lead to atheroprotective or atherogenic responses2. Plexins are key cell-surface receptors of the semaphorin family of cell-guidance signalling proteins and can regulate cellular patterning by modulating the cytoskeleton and focal adhesion structures3-5. However, a role for plexin proteins in mechanotransduction has not been examined. Here we show that plexin D1 (PLXND1) has a role in mechanosensation and mechanically induced disease pathogenesis. PLXND1 is required for the response of endothelial cells to shear stress in vitro and in vivo and regulates the site-specific distribution of atherosclerotic lesions. In endothelial cells, PLXND1 is a direct force sensor and forms a mechanocomplex with neuropilin-1 and VEGFR2 that is necessary and sufficient for conferring mechanosensitivity upstream of the junctional complex and integrins. PLXND1 achieves its binary functions as either a ligand or a force receptor by adopting two distinct molecular conformations. Our results establish a previously undescribed mechanosensor in endothelial cells that regulates cardiovascular pathophysiology, and provide a mechanism by which a single receptor can exhibit a binary biochemical nature.


Asunto(s)
Células Endoteliales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mecanotransducción Celular , Glicoproteínas de Membrana/metabolismo , Estrés Mecánico , Animales , Aterosclerosis/metabolismo , Femenino , Integrinas/metabolismo , Ratones , Neuropilina-1/metabolismo , Docilidad , Receptores de Superficie Celular/metabolismo , Semaforinas/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Circulation ; 139(13): 1612-1628, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30586761

RESUMEN

BACKGROUND: Angiogenesis and vascular remodeling are complementary, innate responses to ischemic cardiovascular events, including peripheral artery disease and myocardial infarction, which restore tissue blood supply and oxygenation; the endothelium plays a critical function in these intrinsic protective processes. C-type natriuretic peptide (CNP) is a fundamental endothelial signaling species that coordinates vascular homeostasis. Herein, we sought to delineate a central role for CNP in angiogenesis and vascular remodeling in response to ischemia. METHODS: The in vitro angiogenic capacity of CNP was examined in pulmonary microvascular endothelial cells and aortic rings isolated from wild-type, endothelium-specific CNP-/-, global natriuretic peptide receptor (NPR)-B-/- and NPR-C-/- animals, and human umbilical vein endothelial cells. These studies were complemented by in vivo investigation of neovascularization and vascular remodeling after ischemia or vessel injury, and CNP/NPR-C expression and localization in tissue from patients with peripheral artery disease. RESULTS: Clinical vascular ischemia is associated with reduced levels of CNP and its cognate NPR-C. Moreover, genetic or pharmacological inhibition of CNP and NPR-C, but not NPR-B, reduces the angiogenic potential of pulmonary microvascular endothelial cells, human umbilical vein endothelial cells, and isolated vessels ex vivo. Angiogenesis and remodeling are impaired in vivo in endothelium-specific CNP-/- and NPR-C-/-, but not NPR-B-/-, mice; the detrimental phenotype caused by genetic deletion of endothelial CNP, but not NPR-C, can be rescued by pharmacological administration of CNP. The proangiogenic effect of CNP/NPR-C is dependent on activation of Gi, ERK1/2, and phosphoinositide 3-kinase γ/Akt at a molecular level. CONCLUSIONS: These data define a central (patho)physiological role for CNP in angiogenesis and vascular remodeling in response to ischemia and provide the rationale for pharmacological activation of NPR-C as an innovative approach to treating peripheral artery disease and ischemic cardiovascular disorders.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Péptido Natriurético Tipo-C/metabolismo , Neovascularización Fisiológica , Transducción de Señal , Animales , Hipoxia de la Célula , Humanos , Ratones , Ratones Noqueados , Péptido Natriurético Tipo-C/genética , Remodelación Vascular
4.
Am J Physiol Cell Physiol ; 316(3): C424-C433, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30649916

RESUMEN

Neuropilin 1 (NRP1) is important for neuronal and cardiovascular development due to its role in conveying class 3 semaphorin and vascular endothelial growth factor signaling, respectively. NRP1 is expressed in smooth muscle cells (SMCs) and mediates their migration and proliferation in cell culture and is implicated in pathological SMC remodeling in vivo. To address the importance of Nrp1 for SMC function during development, we generated conditional inducible Nrp1 SMC-specific knockout mice. Induction of early postnatal SMC-specific Nrp1 knockout led to pulmonary hemorrhage associated with defects in alveogenesis and revealed a specific requirement for Nrp1 in myofibroblast recruitment to the alveolar septae and PDGF-AA-induced migration in vitro. Furthermore, SMC-specific Nrp1 knockout inhibited PDGF-BB-stimulated SMC outgrowth ex vivo in aortic ring assays and reduced pathological arterial neointima formation in vivo. In contrast, we observed little significant effect of SMC-specific Nrp1 knockout on neonatal retinal vascularization. Our results point to a requirement of Nrp1 in vascular smooth muscle and myofibroblast function in vivo, which may have relevance for postnatal lung development and for pathologies characterized by excessive SMC and/or myofibroblast proliferation.


Asunto(s)
Pulmón/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neuropilina-1/metabolismo , Animales , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Neointima/metabolismo , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 38(8): 1845-1858, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29880492

RESUMEN

Objective- NRP1(neuropilin-1) acts as a coreceptor for VEGF (vascular endothelial growth factor) with an essential role in angiogenesis. Recent findings suggest that posttranslational proteolytic cleavage of VEGF receptors may be an important mechanism for regulating angiogenesis, but the role of NRP1 proteolysis and the NRP1 species generated by cleavage in endothelial cells is not known. Here, we characterize NRP1 proteolytic cleavage in endothelial cells, determine the mechanism, and investigate the role of NRP1 cleavage in regulation of endothelial cell function. Approach and Results- NRP1 species comprising the carboxy (C)-terminal and transmembrane NRP1 domains but lacking the ligand-binding A and B regions are constitutively expressed in endothelial cells. Generation of these C-terminal domain NRP1 proteins is upregulated by phorbol ester and Ca2+ ionophore, and reduced by pharmacological inhibition of metalloproteinases, by small interfering RNA-mediated knockdown of 2 members of ADAM (a disintegrin and metalloproteinase) family, ADAMs 9 and 10, and by a specific ADAM10 inhibitor. Furthermore, VEGF upregulates expression of these NRP1 species in an ADAM9/10-dependent manner. Transduction of endothelial cells with adenoviral constructs expressing NRP1 C-terminal domain fragments inhibited VEGF-induced phosphorylation of VEGFR2 (VEGF receptor tyrosine kinase)/KDR (kinase domain insert receptor) and decreased VEGF-stimulated endothelial cell motility and angiogenesis in coculture and aortic ring sprouting assays. Conclusions- These findings identify novel NRP1 species in endothelial cells and demonstrate that regulation of NRP1 proteolysis via ADAMs 9 and 10 is a new regulatory pathway able to modulate VEGF angiogenic signaling.


Asunto(s)
Proteínas ADAMTS/metabolismo , Proteína ADAMTS9/metabolismo , Inductores de la Angiogénesis/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Neuropilina-1/metabolismo , Fragmentos de Péptidos/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Proteínas ADAMTS/genética , Proteína ADAMTS9/genética , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Ratones , Neuropilina-1/genética , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Transducción de Señal/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Nature ; 540(7634): 531-532, 2016 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-27926734
7.
J Cell Sci ; 127(Pt 12): 2647-58, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24762811

RESUMEN

DOK1 regulates platelet-derived growth factor (PDGF)-BB-stimulated glioma cell motility. Mechanisms regulating tumour cell motility are essential for invasion and metastasis. We report here that PDGF-BB-mediated glioma cell invasion and migration are dependent on the adaptor protein downstream of kinase 1 (DOK1). DOK1 is expressed in several glioma cell lines and in tumour biopsies from high-grade gliomas. DOK1 becomes tyrosine phosphorylated upon PDGF-BB stimulation of human glioma cells. Knockdown of DOK1 or expression of a DOK1 mutant (DOK1FF) containing Phe in place of Tyr at residues 362 and 398, resulted in inhibition of both the PDGF-BB-induced tyrosine phosphorylation of p130Cas (also known as BCAR1) and the activation of Rap1. DOK1 colocalises with tyrosine phosphorylated p130Cas at the cell membrane of PDGF-BB-treated cells. Expression of a non-tyrosine-phosphorylatable substrate domain mutant of p130Cas (p130Cas15F) inhibited PDGF-BB-mediated Rap1 activation. Knockdown of DOK1 and Rap1 inhibited PDGF-BB-induced chemotactic cell migration, and knockdown of DOK1 and Rap1 and expression of DOK1FF inhibited PDGF-mediated three-dimensional (3D) spheroid invasion. These data show a crucial role for DOK1 in the regulation of PDGF-BB-mediated tumour cell motility through a p130Cas-Rap1 signalling pathway. [Corrected]


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteína Sustrato Asociada a CrK/metabolismo , Proteínas de Unión al ADN/fisiología , Glioblastoma/metabolismo , Fosfoproteínas/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Proteínas de Unión al ARN/fisiología , Proteínas de Unión a Telómeros/metabolismo , Becaplermina , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Quimiotaxis , Glioblastoma/patología , Humanos , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Complejo Shelterina , Transducción de Señal , Familia-src Quinasas/metabolismo
8.
Methods Mol Biol ; 2475: 229-238, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35451762

RESUMEN

The ex vivo aortic ring assay is one of the most widely used protocols to study sprouting angiogenesis. It is a highly adaptable method that can be utilized to investigate the effects of different growth factors, small-molecule drugs, and genetic modifications on vascular sprouting in a physiologically relevant setting. In this chapter we describe a simple and optimized protocol for investigating vascular sprouting in the mouse aortic ring model. The protocol describes the harvesting and embedding of the aortic rings in a collagen matrix, treatment of the rings with agents of interest, and the visualization and quantification of the vascular sprouts.


Asunto(s)
Vasos Linfáticos , Neovascularización Fisiológica , Adventicia , Animales , Aorta , Ratones , Neovascularización Patológica , Neovascularización Fisiológica/fisiología
9.
J Cell Biol ; 221(2)2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35024764

RESUMEN

The repertoire of extratranslational functions of components of the protein synthesis apparatus is expanding to include control of key cell signaling networks. However, very little is known about noncanonical functions of members of the protein synthesis machinery in regulating cellular mechanics. We demonstrate that the eukaryotic initiation factor 6 (eIF6) modulates cellular mechanobiology. eIF6-depleted endothelial cells, under basal conditions, exhibit unchanged nascent protein synthesis, polysome profiles, and cytoskeleton protein expression, with minimal effects on ribosomal biogenesis. In contrast, using traction force and atomic force microscopy, we show that loss of eIF6 leads to reduced stiffness and force generation accompanied by cytoskeletal and focal adhesion defects. Mechanistically, we show that eIF6 is required for the correct spatial mechanoactivation of ERK1/2 via stabilization of an eIF6-RACK1-ERK1/2-FAK mechanocomplex, which is necessary for force-induced remodeling. These results reveal an extratranslational function for eIF6 and a novel paradigm for how mechanotransduction, the cellular cytoskeleton, and protein translation constituents are linked.


Asunto(s)
Células Endoteliales/metabolismo , Mecanotransducción Celular , Factores de Iniciación de Péptidos/metabolismo , Animales , Fenómenos Biomecánicos , Bovinos , Citoesqueleto/metabolismo , Adhesiones Focales/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Biosíntesis de Proteínas , Ribosomas/metabolismo
10.
Prenat Diagn ; 31(7): 720-34, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21618255

RESUMEN

Prenatal gene therapy aims to deliver genes to cells and tissues early in prenatal life, allowing correction of a genetic defect, before long-term tissue damage has occurred. In contrast to postnatal gene therapy, prenatal application can target genes to a large population of dividing stem cells, and the smaller fetal size allows a higher vector-to-target cell ratio to be achieved. Early-gestation delivery may allow the development of immune tolerance to the transgenic protein which would facilitate postnatal repeat vector administration if needed. Targeting particular organs will depend on manipulating the vector to achieve selective tropism and on choosing the most appropriate gestational age and injection method for fetal delivery. Intra-amniotic injection reaches the skin, and other organs that are bathed in the fluid however since gene transfer to the lung and gut is usually poor more direct injection methods will be needed. Delivery to the liver and blood can be achieved by systemic delivery via the umbilical vein or peritoneal cavity. Gene transfer to the central nervous system in the fetus is difficult but newer vectors are available that transduce neuronal tissue even after systemic delivery.


Asunto(s)
Terapias Fetales/métodos , Terapias Fetales/tendencias , Enfermedades Genéticas Congénitas/terapia , Terapia Genética/métodos , Animales , Células Madre Embrionarias/trasplante , Femenino , Terapias Fetales/efectos adversos , Técnicas de Transferencia de Gen/efectos adversos , Terapia Genética/efectos adversos , Terapia Genética/tendencias , Humanos , Especificidad de Órganos/genética , Embarazo
11.
Sci Adv ; 7(28)2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34244146

RESUMEN

The response of endothelial cells to mechanical forces is a critical determinant of vascular health. Vascular pathologies, such as atherosclerosis, characterized by abnormal mechanical forces are frequently accompanied by endothelial-to-mesenchymal transition (EndMT). However, how forces affect the mechanotransduction pathways controlling cellular plasticity, inflammation, and, ultimately, vessel pathology is poorly understood. Here, we identify a mechanoreceptor that is sui generis for EndMT and unveil a molecular Alk5-Shc pathway that leads to EndMT and atherosclerosis. Depletion of Alk5 abrogates shear stress-induced EndMT responses, and genetic targeting of endothelial Shc reduces EndMT and atherosclerosis in areas of disturbed flow. Tensional force and reconstitution experiments reveal a mechanosensory function for Alk5 in EndMT signaling that is unique and independent of other mechanosensors. Our findings are of fundamental importance for understanding how mechanical forces regulate biochemical signaling, cell plasticity, and vascular disease.

12.
Hum Gene Ther ; 31(21-22): 1190-1202, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32988220

RESUMEN

Severe fetal growth restriction (FGR) affects 1:500 pregnancies, is untreatable and causes serious neonatal morbidity and death. Reduced uterine blood flow (UBF) and lack of bioavailable VEGF due to placental insufficiency is a major cause. Transduction of uterine arteries in normal or FGR sheep and guinea pigs using an adenovirus (Ad) encoding VEGF isoforms A (Ad.VEGF-A165) and a FLAG-tagged pre-processed short form D (DΔNΔC, Ad.VEGF-DΔNΔC-FLAG) increases endothelial nitric oxide expression, enhances relaxation and reduces constriction of the uterine arteries and their branches. UBF and angiogenesis are increased long term, improving fetal growth in utero. For clinical trial development we compared Ad.VEGF vector transduction efficiency and function in endothelial cells (ECs) derived from different species. We aimed to compare the transduction efficiency and function of the pre-clinical study Ad. constructs (Ad.VEGF-A165, Ad.VEGF-DΔNΔC-FLAG) with the intended clinical trial construct (Ad.VEGF-DΔNΔC) where the FLAG tag is removed. We infected ECs from human umbilical vein, pregnant sheep uterine artery, pregnant guinea pig aorta and non-pregnant rabbit aorta, with increasing multiplicity of infection (MOI) for 24 or 48 hours of three Ad.VEGF vectors, compared to control Ad. containing the LacZ gene (Ad.LacZ). VEGF supernatant expression was analysed by ELISA. Functional assessment used tube formation assay and Erk-Akt phosphorylation by ELISA. VEGF expression was higher after Ad.VEGF-DΔNΔC-FLAG and Ad.VEGF-DΔNΔC transduction compared to Ad.VEGF-A165 in all EC types (*p < 0.001). Tube formation was higher in ECs transduced with Ad.VEGF-DΔNΔC in all species compared to other constructs (***p < 0.001, *p < 0.05 with rabbit aortic ECs). Phospho-Erk and phospho-Akt assays displayed no differences between the three vector constructs, whose effect was, as in other experiments, higher than Ad.LacZ (***p < 0.001). In conclusion, we observed high transduction efficiency and functional effects of Ad.VEGF-DΔNΔC vector with comparability in major pathway activation to constructs used in pre-clinical studies, supporting its use in a clinical trial.


Asunto(s)
Adenoviridae/genética , Dependovirus/genética , Endotelio Vascular/metabolismo , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Insuficiencia Placentaria/terapia , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Femenino , Vectores Genéticos/genética , Cobayas , Células Endoteliales de la Vena Umbilical Humana , Humanos , Insuficiencia Placentaria/genética , Insuficiencia Placentaria/metabolismo , Insuficiencia Placentaria/patología , Embarazo , Conejos , Ovinos
13.
Cardiovasc Res ; 116(11): 1863-1874, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31584065

RESUMEN

AIMS: Genome-wide association studies (GWAS) have consistently identified an association between coronary artery disease (CAD) and a locus on chromosome 10 containing a single gene, JCAD (formerly KIAA1462). However, little is known about the mechanism by which JCAD could influence the development of atherosclerosis. METHODS AND RESULTS: Vascular function was quantified in subjects with CAD by flow-mediated dilatation (FMD) and vasorelaxation responses in isolated blood vessel segments. The JCAD risk allele identified by GWAS was associated with reduced FMD and reduced endothelial-dependent relaxations. To study the impact of loss of Jcad on atherosclerosis, Jcad-/- mice were crossed to an ApoE-/- background and fed a high-fat diet from 6 to16 weeks of age. Loss of Jcad did not affect blood pressure or heart rate. However, Jcad-/-ApoE-/- mice developed significantly less atherosclerosis in the aortic root and the inner curvature of the aortic arch. En face analysis revealed a striking reduction in pro-inflammatory adhesion molecules at sites of disturbed flow on the endothelial cell layer of Jcad-/- mice. Loss of Jcad lead to a reduced recovery perfusion in response to hind limb ischaemia, a model of altered in vivo flow. Knock down of JCAD using siRNA in primary human aortic endothelial cells significantly reduced the response to acute onset of flow, as evidenced by reduced phosphorylation of NF-КB, eNOS, and Akt. CONCLUSION: The novel CAD gene JCAD promotes atherosclerotic plaque formation via a role in the endothelial cell shear stress mechanotransduction pathway.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Moléculas de Adhesión Celular/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Circulación Coronaria , Endotelio Vascular/metabolismo , Miembro Posterior/irrigación sanguínea , Mecanotransducción Celular , Animales , Aorta/metabolismo , Aorta/fisiopatología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/fisiopatología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/genética , Aterosclerosis/fisiopatología , Aterosclerosis/prevención & control , Moléculas de Adhesión Celular/genética , Células Cultivadas , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/fisiopatología , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Endotelio Vascular/fisiopatología , Estudio de Asociación del Genoma Completo , Humanos , Isquemia/genética , Isquemia/metabolismo , Isquemia/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Placa Aterosclerótica , Proteínas Proto-Oncogénicas c-akt , Estrés Mecánico
14.
Reprod Sci ; 23(8): 1087-95, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26865541

RESUMEN

Our study aimed to target adenoviral gene therapy to the uteroplacental circulation of pregnant guinea pigs in order to develop a novel therapy for fetal growth restriction. Four methods of delivery of an adenovirus encoding ß-galactosidase (Ad.LacZ) were evaluated: intravascular injection using phosphate-buffered saline (PBS) into (1) uterine artery (UtA) or (2) internal iliac artery or external administration in (3) PBS or (4) pluronic F-127 gel (Sigma Aldrich). Postmortem examination was performed 4 to 7 days after gene transfer. Tissue transduction was assessed by X-gal histochemistry and enzyme-linked immunosorbent assay. External vascular application of the adenovirus vector in combination with pluronic gel had 91.7% success rate in terms of administration (85% maternal survival) and gave the best results for maternal/fetal survival and local transduction efficiency without any spread to maternal or fetal tissues. This study suggests an optimal method of gene delivery to the UtAs of a small rodent for preclinical studies.


Asunto(s)
Retardo del Crecimiento Fetal/terapia , Marcación de Gen/métodos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Circulación Placentaria , Adenoviridae/fisiología , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Cobayas , Arteria Ilíaca , Embarazo , Arteria Radial , Arteria Uterina , beta-Galactosidasa/análisis , beta-Galactosidasa/genética
15.
Hum Gene Ther ; 27(12): 997-1007, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27530140

RESUMEN

In a model of growth-restricted sheep pregnancy, it was previously demonstrated that transient uterine artery VEGF overexpression can improve fetal growth. This approach was tested in guinea-pig pregnancies, where placental physiology is more similar to humans. Fetal growth restriction (FGR) was attained through peri-conceptual nutrient restriction in virgin guinea pigs. Ad.VEGF-A165 or Ad.LacZ (1 × 1010vp) was applied at mid-gestation via laparotomy, delivered externally to the uterine circulation with thermosensitive gel. At short-term (3-8 days post surgery) or at term gestation, pups were weighed, and tissues were sampled for vector spread analysis, VEGF expression, and its downstream effects. Fetal weight at term was increased (88.01 ± 13.36 g; n = 26) in Ad.VEGF-A165-treated animals compared with Ad.LacZ-treated animals (85.52 ± 13.00 g; n = 19; p = 0.028). The brain, liver, and lung weight and crown rump length were significantly larger in short-term analyses, as well as VEGF expression in transduced tissues. At term, molecular analyses confirmed the presence of VEGF transgene in target tissues but not in fetal samples. Tissue histology analysis and blood biochemistry/hematological examination were comparable with controls. Uterine artery relaxation in Ad.VEGF-A165-treated dams was higher compared with Ad.LacZ-treated dams. Maternal uterine artery Ad.VEGF-A165 increases fetal growth velocity and term fetal weight in growth-restricted guinea-pig pregnancy.


Asunto(s)
Adenoviridae/genética , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/terapia , Peso Fetal/genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Femenino , Cobayas , Embarazo , Flujo Sanguíneo Regional
16.
Cell Transplant ; 25(3): 615, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-28836829

RESUMEN

Long-term engraftment and phenotype correction has been difficult to achieve in humans after in utero stem cell transplantation mainly because of allogeneic rejection. Autologous cells could be obtained during gestation from the amniotic fluid with minimal risk for the fetus and the mother. Using a sheep model, we explored the possibility of using amniotic fluid mesenchymal stem cells (AFMSCs) for autologous in utero stem cell/gene therapy. We collected amniotic fluid (AF) under ultrasound-guided amniocentesis in early gestation pregnant sheep ( n = 9, 58 days of gestation, term = 145 days). AFMSCs were isolated and expanded in all sampled fetal sheep. Those cells were transduced using an HIV vector encoding enhanced green fluorescent protein (GFP) with 63.2% (range 38.3-96.2%) transduction efficiency rate. After expansion, transduced AFMSCs were injected into the peritoneal cavity of each donor fetal sheep at 76 days under ultrasound guidance. One ewe miscarried twin fetuses after amniocentesis. Intraperitoneal injection was successful in the remaining 7 fetal sheep giving a 78% survival for the full procedure. Tissues were sampled at postmortem examination 2 weeks later. PCR analysis detected GFP-positive cells in fetal tissues including liver, heart, placenta, membrane, umbilical cord, adrenal gland, and muscle. GFP protein was detected in these tissues by Western blotting and further confirmed by cytofluorimetric and immunofluorescence analyses. This is the first demonstration of autologous stem cell transplantation in the fetus using AFMSCs. Autologous cells derived from AF showed widespread organ migration and could offer an alternative way to ameliorate prenatal congenital disease.

17.
Methods Mol Biol ; 1332: 189-96, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26285755

RESUMEN

In this chapter, we describe a novel method of adenoviral gene transfer to the uterine and radial arteries of pregnant guinea pigs to improve fetal growth. Adenoviruses encoding VEGF-A165 or a reporter gene ß-galactosidase were reconstituted in pluronic gel and applied topically to the exposed uterine and radial arteries following laparotomy. Pluronic gel is a thermosensitive gel that is liquid at 4 °C, but becomes solid as soon as it comes in contact with body temperature. It thereby acts as a slow-release vehicle for viral vectors to the target tissue and also facilitates closer contact of the viruses with the host tissue. Our studies have shown that adenoviral gene delivery using pluronic gel resulted in the highest transduction efficiency compared to intra-arterial administration or external administration in PBS as a vehicle, when measured by X-gal staining, immunohistochemistry, or Western blotting.


Asunto(s)
Desarrollo Fetal/genética , Técnicas de Transferencia de Gen , Terapia Genética , Circulación Placentaria , Transducción Genética , Factor A de Crecimiento Endotelial Vascular/genética , Adenoviridae/genética , Animales , Femenino , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Cobayas , Modelos Animales , Embarazo , Arteria Uterina/metabolismo
18.
Methods Mol Biol ; 1332: 197-204, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26285756

RESUMEN

In this chapter, we describe a safe and effective approach to achieve local VEGF gene transfer to the uterine arteries in pregnant sheep using direct injection of viral vectors into the uterine arteries. This approach resulted in improved fetal growth in growth-restricted pregnancies. Adenoviral vectors encoding VEGF-A165 or a reporter gene ß-galactosidase were dissolved in 10 mL normal saline shortly before administration. A midline laparotomy was performed and the course of the uterine artery identified. The main trunk (just prior to the first bifurcation) was mobilized by dissection and a vessel loop placed beneath it in order to elevate the artery, which was then occluded digitally just proximal to the planned injection site. The adenoviral solution was slowly injected over 1 min, and the occlusion was maintained for a further 4 min to maximize transduction of the downstream endothelium. After ensuring hemostasis, the abdomen was closed in layers.


Asunto(s)
Desarrollo Fetal/genética , Técnicas de Transferencia de Gen , Terapia Genética , Circulación Placentaria , Transducción Genética , Factor A de Crecimiento Endotelial Vascular/genética , Adenoviridae/genética , Animales , Femenino , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Modelos Animales , Embarazo , Ovinos , Arteria Uterina/metabolismo
19.
Cardiovasc Res ; 108(2): 288-98, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26410366

RESUMEN

AIMS: Neuropilins 1 and 2 (NRP1 and NRP2) play crucial roles in endothelial cell migration contributing to angiogenesis and vascular development. Both NRPs are also expressed by cultured vascular smooth muscle cells (VSMCs) and are implicated in VSMC migration stimulated by PDGF-BB, but it is unknown whether NRPs are relevant for VSMC function in vivo. We investigated the role of NRPs in the rat carotid balloon injury model, in which endothelial denudation and arterial stretch induce neointimal hyperplasia involving VSMC migration and proliferation. METHODS AND RESULTS: NRP1 and NRP2 mRNAs and proteins increased significantly following arterial injury, and immunofluorescent staining revealed neointimal NRP expression. Down-regulation of NRP1 and NRP2 using shRNA significantly reduced neointimal hyperplasia following injury. Furthermore, inhibition of NRP1 by adenovirally overexpressing a loss-of-function NRP1 mutant lacking the cytoplasmic domain (ΔC) reduced neointimal hyperplasia, whereas wild-type (WT) NRP1 had no effect. NRP-targeted shRNAs impaired, while overexpression of NRP1 WT and NRP1 ΔC enhanced, arterial re-endothelialization 14 days after injury. Knockdown of either NRP1 or NRP2 inhibited PDGF-BB-induced rat VSMC migration, whereas knockdown of NRP2, but not NRP1, reduced proliferation of cultured rat VSMC and neointimal VSMC in vivo. NRP knockdown also reduced the phosphorylation of PDGFα and PDGFß receptors in rat VSMC, which mediate VSMC migration and proliferation. CONCLUSION: NRP1 and NRP2 play important roles in the regulation of neointimal hyperplasia in vivo by modulating VSMC migration (via NRP1 and NRP2) and proliferation (via NRP2), independently of the role of NRPs in re-endothelialization.


Asunto(s)
Traumatismos de las Arterias Carótidas/metabolismo , Neointima/metabolismo , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo , Angioplastia de Balón/efectos adversos , Animales , Traumatismos de las Arterias Carótidas/etiología , Traumatismos de las Arterias Carótidas/patología , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Endotelio Vascular/fisiología , Hiperplasia , Masculino , Miocitos del Músculo Liso/metabolismo , Neointima/etiología , Neointima/patología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Ratas Sprague-Dawley , Regulación hacia Arriba
20.
Hum Gene Ther ; 25(4): 375-84, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24593228

RESUMEN

Fetal growth restriction (FGR) occurs in ∼8% of pregnancies and is a major cause of perinatal mortality and morbidity. There is no effective treatment. FGR is characterized by reduced uterine blood flow (UBF). In normal sheep pregnancies, local uterine artery (UtA) adenovirus (Ad)-mediated overexpression of vascular endothelial growth factor (VEGF) increases UBF. Herein we evaluated Ad.VEGF therapy in the overnourished adolescent ewe, an experimental paradigm in which reduced UBF from midgestation correlates with reduced lamb birthweight near term. Singleton pregnancies were established using embryo transfer in adolescent ewes subsequently offered a high intake (n=45) or control intake (n=12) of a complete diet to generate FGR or normal fetoplacental growth, respectively. High-intake ewes were randomized midgestation to receive bilateral UtA injections of 5×10¹¹ particles Ad.VEGF-A165 (n=18), control vector Ad.LacZ (n=14), or control saline (n=13). Fetal growth/well-being were evaluated using serial ultrasound. UBF was monitored using indwelling flowprobes until necropsy at 0.9 gestation. Vasorelaxation, neovascularization within the perivascular adventitia, and placental mRNA expression of angiogenic factors/receptors were examined using organ bath analysis, anti-vWF immunohistochemistry, and qRT-PCR, respectively. Ad.VEGF significantly increased ultrasonographic fetal growth velocity at 3-4 weeks postinjection (p=0.016-0.047). At 0.9 gestation fewer fetuses were markedly growth-restricted (birthweight >2SD below contemporaneous control-intake mean) after Ad.VEGF therapy. There was also evidence of mitigated fetal brain sparing (lower biparietal diameter-to-abdominal circumference and brain-to-liver weight ratios). No effects were observed on UBF or neovascularization; however, Ad.VEGF-transduced vessels demonstrated strikingly enhanced vasorelaxation. Placental efficiency (fetal-to-placental weight ratio) and FLT1/KDR mRNA expression were increased in the maternal but not fetal placental compartments, suggesting downstream effects on placental function. Ad.VEGF gene therapy improves fetal growth in a sheep model of FGR, although the precise mechanism of action remains unclear.


Asunto(s)
Adenoviridae/genética , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/terapia , Vectores Genéticos/genética , Placenta/metabolismo , Útero/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Femenino , Retardo del Crecimiento Fetal/diagnóstico por imagen , Expresión Génica , Terapia Genética , Vectores Genéticos/administración & dosificación , Neovascularización Fisiológica , Circulación Placentaria , Embarazo , Flujo Sanguíneo Regional , Transducción Genética , Ultrasonografía , Arteria Uterina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA