Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circ Res ; 123(12): e51-e64, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30566041

RESUMEN

RATIONALE: Pulmonary arterial hypertension is a deadly disease of the pulmonary vasculature for which no disease-modifying therapies exist. Small-vessel stiffening and remodeling are fundamental pathological features of pulmonary arterial hypertension that occur early and drive further endovascular cell dysfunction. Bone marrow (BM)-derived proangiogenic cells (PACs), a specialized heterogeneous subpopulation of myeloid lineage cells, are thought to play an important role in pathogenesis. OBJECTIVE: To determine whether BM-derived PACs directly contributed to experimental pulmonary hypertension (PH) by promoting small-vessel stiffening through 5-HT2B (serotonin 2B receptor)-mediated signaling. METHODS AND RESULTS: We performed BM transplants using transgenic donor animals expressing diphtheria toxin secondary to activation of an endothelial-specific tamoxifen-inducible Cre and induced experimental PH using hypoxia with SU5416 to enhance endovascular injury and ablated BM-derived PACs, after which we measured right ventricular systolic pressures in a closed-chest procedure. BM-derived PAC lineage tracing was accomplished by transplanting BM from transgenic donor animals with fluorescently labeled hematopoietic cells and treating mice with a 5-HT2B antagonist. BM-derived PAC ablation both prevented and reversed experimental PH with SU5416-enhanced endovascular injury, reducing the number of muscularized pulmonary arterioles and normalizing arteriole stiffness as measured by atomic force microscopy. Similarly, treatment with a pharmacological antagonist of 5-HT2B also prevented experimental PH, reducing the number and stiffness of muscularized pulmonary arterioles. PACs accelerated pulmonary microvascular endothelial cell injury response in vitro, and the presence of BM-derived PACs significantly correlated with stiffer pulmonary arterioles in pulmonary arterial hypertension patients and mice with experimental PH. RNA sequencing of BM-derived PACs showed that 5-HT2B antagonism significantly altered biologic pathways regulating cell proliferation, locomotion and migration, and cytokine production and response to cytokine stimulus. CONCLUSIONS: Together, our findings illustrate that BM-derived PACs directly contribute to experimental PH with SU5416-enhanced endovascular injury by mediating small-vessel stiffening and remodeling in a 5-HT2B signaling-dependent manner.


Asunto(s)
Hipertensión Pulmonar/patología , Células Progenitoras Mieloides/metabolismo , Receptor de Serotonina 5-HT2B/metabolismo , Rigidez Vascular , Inhibidores de la Angiogénesis/toxicidad , Animales , Arteriolas/patología , Linaje de la Célula , Células Cultivadas , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/etiología , Indoles/toxicidad , Pulmón/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/trasplante , Pirroles/toxicidad
2.
J Clin Invest ; 127(6): 2262-2276, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28463231

RESUMEN

Pulmonary vascular disease is characterized by remodeling and loss of microvessels and is typically attributed to pathological responses in vascular endothelium or abnormal smooth muscle cell phenotypes. We have challenged this understanding by defining an adult pulmonary mesenchymal progenitor cell (MPC) that regulates both microvascular function and angiogenesis. The current understanding of adult MPCs and their roles in homeostasis versus disease has been limited by a lack of genetic markers with which to lineage label multipotent mesenchyme and trace the differentiation of these MPCs into vascular lineages. Here, we have shown that lineage-labeled lung MPCs expressing the ATP-binding cassette protein ABCG2 (ABCG2+) are pericyte progenitors that participate in microvascular homeostasis as well as adaptive angiogenesis. Activation of Wnt/ß-catenin signaling, either autonomously or downstream of decreased BMP receptor signaling, enhanced ABCG2+ MPC proliferation but suppressed MPC differentiation into a functional pericyte lineage. Thus, enhanced Wnt/ß-catenin signaling in ABCG2+ MPCs drives a phenotype of persistent microvascular dysfunction, abnormal angiogenesis, and subsequent exacerbation of bleomycin-induced fibrosis. ABCG2+ MPCs may, therefore, account in part for the aberrant microvessel function and remodeling that are associated with chronic lung diseases.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Microvasos/fisiopatología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Humanos , Pulmón/irrigación sanguínea , Ratones Transgénicos , Microvasos/patología , Neovascularización Patológica/metabolismo , Pericitos/fisiología , Estabilidad Proteica , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Vasoconstricción , Vía de Señalización Wnt
3.
PLoS One ; 11(2): e0148657, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26863209

RESUMEN

Serotonergic anorexigens are the primary pharmacologic risk factor associated with pulmonary arterial hypertension (PAH), and the resulting PAH is clinically indistinguishable from the heritable form of disease, associated with BMPR2 mutations. Both BMPR2 mutation and agonists to the serotonin receptor HTR2B have been shown to cause activation of SRC tyrosine kinase; conversely, antagonists to HTR2B inhibit SRC trafficking and downstream function. To test the hypothesis that a HTR2B antagonist can prevent BMRP2 mutation induced PAH by restricting aberrant SRC trafficking and downstream activity, we exposed BMPR2 mutant mice, which spontaneously develop PAH, to a HTR2B antagonist, SB204741, to block the SRC activation caused by BMPR2 mutation. SB204741 prevented the development of PAH in BMPR2 mutant mice, reduced recruitment of inflammatory cells to their lungs, and reduced muscularization of their blood vessels. By atomic force microscopy, we determined that BMPR2 mutant mice normally had a doubling of vessel stiffness, which was substantially normalized by HTR2B inhibition. SB204741 reduced SRC phosphorylation and downstream activity in BMPR2 mutant mice. Gene expression arrays indicate that the primary changes were in cytoskeletal and muscle contractility genes. These results were confirmed by gel contraction assays showing that HTR2B inhibition nearly normalizes the 400% increase in gel contraction normally seen in BMPR2 mutant smooth muscle cells. Heritable PAH results from increased SRC activation, cellular contraction, and vascular resistance, but antagonism of HTR2B prevents SRC phosphorylation, downstream activity, and PAH in BMPR2 mutant mice.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Hipertensión Pulmonar/prevención & control , Indoles/farmacología , Receptor de Serotonina 5-HT2B/genética , Antagonistas de la Serotonina/farmacología , Urea/análogos & derivados , Familia-src Quinasas/genética , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/deficiencia , Movimiento Celular/efectos de los fármacos , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Transgénicos , Contracción Muscular/efectos de los fármacos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutación , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Transporte de Proteínas , Receptor de Serotonina 5-HT2B/metabolismo , Transducción de Señal , Urea/farmacología , Rigidez Vascular/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
5.
Am J Physiol Cell Physiol ; 307(5): C415-30, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24871858

RESUMEN

Understanding differences in gene expression that increase risk for pulmonary arterial hypertension (PAH) is essential to understanding the molecular basis for disease. Previous studies on patient samples were limited by end-stage disease effects or by use of nonadherent cells, which are not ideal to model vascular cells in vivo. These studies addressed the hypothesis that pathological processes associated with PAH may be identified via a genetic signature common across multiple cell types. Expression array experiments were initially conducted to analyze cell types at different stages of vascular differentiation (mesenchymal stromal and endothelial) derived from PAH patient-specific induced pluripotent stem (iPS) cells. Molecular pathways that were altered in the PAH cell lines were then compared with those in fibroblasts from 21 patients, including those with idiopathic and heritable PAH. Wnt was identified as a target pathway and was validated in vitro using primary patient mesenchymal and endothelial cells. Taken together, our data suggest that the molecular lesions that cause PAH are present in all cell types evaluated, regardless of origin, and that stimulation of the Wnt signaling pathway was a common molecular defect in both heritable and idiopathic PAH.


Asunto(s)
Diferenciación Celular/genética , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Células Madre Pluripotentes/patología , Vía de Señalización Wnt/genética , Línea Celular , Células Cultivadas , Células Endoteliales/patología , Células Endoteliales/fisiología , Hipertensión Pulmonar Primaria Familiar , Humanos , Células Madre Pluripotentes/fisiología , Mucosa Respiratoria/patología , Mucosa Respiratoria/fisiología
6.
Biotechnol J ; 8(4): 485-95, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23447378

RESUMEN

Recently there has been an increased interest in the effects of paracrine signaling between groups of cells, particularly in the context of better understanding how stem cells contribute to tissue repair. Most current 3D co-culture methods lack the ability to effectively separate two cell populations after the culture period, which is important for simultaneously analyzing the reciprocal effects of each cell type on the other. Here, we detail the development of a 3D hydrogel co-culture system that allows us to culture different cell types for up to 7 days and subsequently separate and isolate the different cell populations using enzyme-sensitive glues. Separable 3D co-culture laminates were prepared by laminating PEG-based hydrogels with enzyme-degradable hydrogel adhesives. Encapsulated cell populations exhibited good segregation with well-defined interfaces. Furthermore, constructs can be separated on-demand upon addition of the appropriate enzyme, while cell viability remains high throughout the culture period, even after laminate separation. This platform offers great potential for a variety of basic cell signaling studies as the incorporation of an enzyme-sensitive adhesive interface allows the on-demand separation of individual cell populations for immediate analysis or further culture to examine persistence of co-culture effects and paracrine signaling on cell populations.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Separación Celular/métodos , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Análisis de Varianza , Técnicas de Cultivo de Célula/métodos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo/instrumentación , Técnicas de Cocultivo/métodos , Diseño de Equipo , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Polietilenglicoles/química
7.
Acta Biomater ; 7(3): 986-95, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20965281

RESUMEN

Size scale plays an important role in the release properties and cellular presentation of drug delivery vehicles. Because negatively charged chondroitin sulfate (CS) is capable of electrostatically sequestering positively charged growth factors, CS-derived nanoscale micelles and microscale spheroids were synthesized as potential growth factor carriers to enhance differentiation of stem cells. Particles were characterized for morphology, size distribution, surface charge and cytocompatibility, as well as release of transforming growth factor-ß1 (TGF-ß1) and tumor necrosis factor-α (TNF-α). CS micelles were spherical and negatively charged with a bimodal distribution of 324.1±8.5 and 73.2±4.4 nm diameters, and CS microspheres possessed a rounded morphology and a diameter of 4.3±0.93 µm. Positively charged TGF-ß1 demonstrated minimal release after loading in CS microspheres, while negatively charged TNF-α exhibited substantial release over the first 15 h, suggesting that TGF-ß1 electrostatically complexed with CS. The micelles and microparticles were found to be cytocompatible at moderate concentrations with marrow stromal cell monolayers and within embryonic stem cell embryoid bodies. These synthesis techniques, which allow the formation of CS-based carriers over a variety of nano- and microscale sizes, offer versatility for tailored release of positively charged growth factors and controlled CS presentation for a variety of stem cell-based applications in tissue engineering and regenerative medicine.


Asunto(s)
Sulfatos de Condroitina/química , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Microesferas , Nanopartículas , Animales , Células Madre Embrionarias/metabolismo , Espectroscopía de Resonancia Magnética , Ratones , Micelas , Electricidad Estática
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...