Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 31(6): 1300-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21415388

RESUMEN

OBJECTIVE: PTEN inactivation selectively in smooth muscle cells (SMC) initiates multiple downstream events driving neointima formation, including SMC cytokine/chemokine production, in particular stromal cell-derived factor-1α (SDF-1α). We investigated the effects of SDF-1α on resident SMC and bone marrow-derived cells and in mediating neointima formation. METHODS AND RESULTS: Inducible, SMC-specific PTEN knockout mice (PTEN iKO) were bred to floxed-stop ROSA26-ß-galactosidase (ßGal) mice to fate-map mature SMC in response to injury; mice received wild-type green fluorescent protein-labeled bone marrow to track recruitment. Following wire-induced femoral artery injury, ßGal(+) SMC accumulated in the intima and adventitia. Compared with wild-type, PTEN iKO mice exhibited massive neointima formation, increased replicating intimal and medial ßGal(+)SMC, and enhanced vascular recruitment of bone marrow cells following injury. Inhibiting SDF-1α blocked these events and reversed enhanced neointima formation observed in PTEN iKO mice. Most recruited green fluorescent protein(+) cells stained positive for macrophage markers but not SMC markers. SMC-macrophage interactions resulted in a persistent SMC inflammatory phenotype that was dependent on SMC PTEN and SDF-1α expression. CONCLUSION: Resident SMC play a multifaceted role in neointima formation by contributing the majority of neointimal cells, regulating recruitment of inflammatory cells, and contributing to adventitial remodeling. The SMC PTEN-SDF-1α axis is a critical regulator of these events.


Asunto(s)
Quimiocina CXCL12/fisiología , Miocitos del Músculo Liso/fisiología , Neointima/etiología , Fosfohidrolasa PTEN/fisiología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Proliferación Celular , Células Madre Hematopoyéticas/citología , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/citología
2.
Cardiovasc Res ; 86(2): 274-82, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20051384

RESUMEN

AIMS: Phosphatase and tensin homolog (PTEN) is implicated as a negative regulator of vascular smooth muscle cell (SMC) proliferation and injury-induced vascular remodelling. We tested if selective depletion of PTEN only in SMC is sufficient to promote SMC phenotypic modulation, cytokine production, and enhanced neointima formation. METHODS AND RESULTS: Smooth muscle marker expression and induction of pro-inflammatory cytokines were compared in cultured SMC expressing control or PTEN-specific shRNA. Compared with controls, PTEN-deficient SMC exhibited increased phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signalling and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappaB) activity, reduced expression of SM markers (SM-alpha-actin and calponin), and increased production of stromal cell-derived factor-1alpha (SDF-1alpha), monocyte chemotactic protein-1 (MCP-1), interleukin-6 (IL-6), and chemokine (C-X-C motif) ligand 1 (KC/CXCL1) under basal conditions. PI3K/Akt or mTOR inhibition reversed repression of SM marker expression, whereas PI3K/Akt or NF-kappaB inhibition blocked cytokine induction mediated by PTEN depletion. Carotid ligation in mice with genetic reduction of PTEN specifically in SMC (SMC-specific PTEN heterozygotes) resulted in enhanced neointima formation, increased SMC hyperplasia, reduced SM-alpha-actin and calponin expression, and increased NF-kappaB and cytokine expression compared with wild-types. Lesion formation in SMC-specific heterozygotes was similar to lesion formation in global PTEN heterozygotes, indicating that inactivation of PTEN exclusively in SMC is sufficient to induce considerable increases in neointima formation. CONCLUSION: PTEN activation specifically in SMC is a common upstream regulator of multiple downstream events involved in pathological vascular remodelling, including proliferation, de-differentiation, and production of multiple cytokines.


Asunto(s)
Traumatismos de las Arterias Carótidas/enzimología , Proliferación Celular , Inflamación/enzimología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Fosfohidrolasa PTEN/deficiencia , Túnica Íntima/enzimología , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/inmunología , Traumatismos de las Arterias Carótidas/patología , Desdiferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Noqueados , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/patología , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/genética , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Ratas , Transducción de Señal , Serina-Treonina Quinasas TOR , Túnica Íntima/inmunología , Túnica Íntima/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...