Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Perfusion ; 37(6): 582-589, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-33899586

RESUMEN

Vascular endothelial cell stimulation is associated with the activation of different signalling pathways and transcription factors. Acute shear stress is known to induce different pro-inflammatory mediators such as IL-8. Nrf2 is activated by prolonged high shear stress promoting an antiinflammatory and athero-protective environment. However, little is known about the impact of acute shear stress on Nrf2 and Keap1 function and its role in IL-8 regulation. We aimed to examine Nrf2-Keap1 complex activation in-vitro and its role in regulating IL-8 transcripts under acute arterial shear stress (12 dyn/cm2) in venous endothelial cells (ECs). We note that acute high shear stress caused a significant upregulation of Nrf2 target genes, HO-1 and GCLM and an increased IL-8 upregulation at 90 and 120 minutes. Mechanistically, acute high shear did not affect Nrf2 nuclear translocation but resulted in reduced nuclear Keap1, suggesting that the reduction in nuclear Keap1 may result in increased free nuclear nrf2 to induce transcription. Consistently, the suppression of Keap1 using shRNA (shKeap1) resulted in significant upregulation of IL-8 transcripts in response to acute shear stress. Interestingly; the over expression of Nrf2 using Nrf2-Ad-WT or Sulforaphane was also associated with significant upregulation of IL-8 compared to controls. This study highlights the role of Keap1 in Nrf2 activation under shear stress and indicates that activation of Nrf2 may be deleterious in ECs in the context of acute haemodynamic injury.


Asunto(s)
Células Endoteliales , Factor 2 Relacionado con NF-E2 , Células Endoteliales/metabolismo , Humanos , Interleucina-8/genética , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/fisiología , Estrés Mecánico
2.
Diabetologia ; 62(7): 1297-1311, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31016359

RESUMEN

AIMS/HYPOTHESIS: Sensory neuropathy is common in people with diabetes; neuropathy can also affect the bone marrow of individuals with type 2 diabetes. However, no information exists on the state of bone marrow sensory innervation in type 1 diabetes. Sensory neurons are trophically dependent on nerve growth factor (NGF) for their survival. The aim of this investigation was twofold: (1) to determine if sensory neuropathy affects the bone marrow in a mouse model of type 1 diabetes, with consequences for stem cell liberation after tissue injury; and (2) to verify if a single systemic injection of the NGF gene exerts long-term beneficial effects on these phenomena. METHODS: A mouse model of type 1 diabetes was generated in CD1 mice by administration of streptozotocin; vehicle was administered to non-diabetic control animals. Diabetic animals were randomised to receive systemic gene therapy with either human NGF or ß-galactosidase. After 13 weeks, limb ischaemia was induced in both groups to study the recovery post injury. When the animals were killed, samples of tissue and peripheral blood were taken to assess stem cell mobilisation and homing, levels of substance P and muscle vascularisation. An in vitro cellular model was adopted to verify signalling downstream to human NGF and related neurotrophic or pro-apoptotic effects. Normally distributed variables were compared between groups using the unpaired Student's t test and non-normally distributed variables were assessed by the Wilcoxon-Mann-Whitney test. The Fisher's exact test was employed for categorical variables. RESULTS: Immunohistochemistry indicated a 3.3-fold reduction in the number of substance P-positive nociceptive fibres in the bone marrow of type 1 diabetic mice (p < 0.001 vs non-diabetic). Moreover, diabetes abrogated the creation of a neurokinin gradient which, in non-diabetic mice, favoured the mobilisation and homing of bone-marrow-derived stem cells expressing the substance P receptor neurokinin 1 receptor (NK1R). Pre-emptive gene therapy with NGF prevented bone marrow denervation, contrasting with the inhibitory effect of diabetes on the mobilisation of NK1R-expressing stem cells, and restored blood flow recovery from limb ischaemia. In vitro hNGF induced neurite outgrowth and exerted anti-apoptotic actions on rat PC12 cells exposed to high glucose via activation of the canonical neurotrophic tyrosine kinase receptor type 1 (TrkA) signalling pathway. CONCLUSIONS/INTERPRETATION: This study shows, for the first time, the occurrence of sensory neuropathy in the bone marrow of type 1 diabetic mice, which translates into an altered modulation of substance P and depressed release of substance P-responsive stem cells following ischaemia. NGF therapy improves bone marrow sensory innervation, with benefits for healing on the occurrence of peripheral ischaemia. Nociceptors may represent a new target for the treatment of ischaemic complications in diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Terapia Genética/métodos , Factor de Crecimiento Nervioso/metabolismo , Células Receptoras Sensoriales/citología , Células Madre/citología , Animales , Médula Ósea , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/metabolismo , Inmunohistoquímica , Isquemia/terapia , Masculino , Ratones , Células Receptoras Sensoriales/metabolismo , Células Madre/metabolismo
3.
J Mol Cell Cardiol ; 90: 1-10, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26625714

RESUMEN

AIMS: Inhibition of vascular smooth muscle cell (VSMC) proliferation by intracellular cAMP prevents excessive neointima formation and hence angioplasty restenosis and vein-graft failure. These protective effects are mediated via actin-cytoskeleton remodelling and subsequent regulation of gene expression by mechanisms that are incompletely understood. Here we investigated the role of components of the growth-regulatory Hippo pathway, specifically the transcription factor TEAD and its co-factors YAP and TAZ in VSMC. METHODS AND RESULTS: Elevation of cAMP using forskolin, dibutyryl-cAMP or the physiological agonists, Cicaprost or adenosine, significantly increased phosphorylation and nuclear export YAP and TAZ and inhibited TEAD-luciferase report gene activity. Similar effects were obtained by inhibiting RhoA activity with C3-transferase, its downstream kinase, ROCK, with Y27632, or actin-polymerisation with Latrunculin-B. Conversely, expression of constitutively-active RhoA reversed the inhibitory effects of forskolin on TEAD-luciferase. Forskolin significantly inhibited the mRNA expression of the pro-mitogenic genes, CCN1, CTGF, c-MYC and TGFB2 and this was reversed by expression of constitutively-active YAP or TAZ phospho-mutants. Inhibition of YAP and TAZ function with RNAi or Verteporfin significantly reduced VSMC proliferation. Furthermore, the anti-mitogenic effects of forskolin were reversed by overexpression of constitutively-active YAP or TAZ. CONCLUSION: Taken together, these data demonstrate that cAMP-induced actin-cytoskeleton remodelling inhibits YAP/TAZ-TEAD dependent expression of pro-mitogenic genes in VSMC. This mechanism contributes novel insight into the anti-mitogenic effects of cAMP in VSMC and suggests a new target for intervention.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , AMP Cíclico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Miocitos del Músculo Liso/metabolismo , Amidas/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Bucladesina/metabolismo , Bucladesina/farmacología , Proliferación Celular/efectos de los fármacos , Colforsina/farmacología , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Proteína 61 Rica en Cisteína/genética , Proteína 61 Rica en Cisteína/metabolismo , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Tiazolidinas/farmacología , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta2/metabolismo , Proteínas Señalizadoras YAP , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
4.
J Mol Cell Cardiol ; 79: 157-68, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25446180

RESUMEN

Elevation of intracellular cAMP concentration has numerous vascular protective effects that are in part mediated via actin cytoskeleton-remodelling and subsequent regulation of gene expression. However, the mechanisms are incompletely understood. Here we investigated whether cAMP-induced actin-cytoskeleton remodelling modulates VSMC behaviour by inhibiting expression of CCN1. In cultured rat VSMC, CCN1-silencing significantly inhibited BrdU incorporation and migration in a wound healing assay. Recombinant CCN1 enhanced chemotaxis in a Boyden chamber. Adding db-cAMP, or elevating cAMP using forskolin, significantly inhibited CCN1 mRNA and protein expression in vitro; transcriptional regulation was demonstrated by measuring pre-spliced CCN1 mRNA and CCN1-promoter activity. Forskolin also inhibited CCN1 expression in balloon injured rat carotid arteries in vivo. Inhibiting RhoA activity, which regulates actin-polymerisation, by cAMP-elevation or pharmacologically with C3-transferase, or inhibiting its downstream kinase, ROCK, with Y27632, significantly inhibited CCN1 expression. Conversely, expression of constitutively active RhoA reversed the inhibitory effects of forskolin on CCN1 mRNA. Furthermore, CCN1 mRNA levels were significantly decreased by inhibiting actin-polymerisation with latrunculin B or increased by stimulating actin-polymerisation with Jasplakinolide. We next tested the role of the actin-dependent SRF co-factor, MKL1, in CCN1 expression. Forskolin inhibited nuclear translocation of MKL1 and binding of MKL1 to the CCN1 promoter. Constitutively-active MKL1 enhanced basal promoter activity of wild-type but not SRE-mutated CCN1; and prevented forskolin inhibition. Furthermore, pharmacological MKL-inhibition with CCG-1423 significantly inhibited CCN1 promoter activity as well as mRNA and protein expression. Our data demonstrates that cAMP-induced actin-cytoskeleton remodelling regulates expression of CCN1 through MKL1: it highlights a novel cAMP-dependent mechanism controlling VSMC behaviour.


Asunto(s)
Citoesqueleto de Actina/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , AMP Cíclico/farmacología , Proteína 61 Rica en Cisteína/genética , Transactivadores/metabolismo , Adenosina/farmacología , Aminopiridinas/farmacología , Animales , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Proliferación Celular/efectos de los fármacos , Colforsina/farmacología , Proteína 61 Rica en Cisteína/metabolismo , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Humanos , Masculino , Mitógenos/farmacología , Modelos Biológicos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Ratas Sprague-Dawley , Factor de Respuesta Sérica/metabolismo , Factores de Transcripción , Transcripción Genética/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
5.
Mediators Inflamm ; 2014: 276457, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25301980

RESUMEN

Matrix metalloproteinase-14 (MMP-14) promotes vulnerable plaque morphology in mice, whereas tissue inhibitor of metalloproteinases-3 (TIMP-3) overexpression is protective. MMP-14(hi) TIMP-3(lo) rabbit foam cells are more invasive and more prone to apoptosis than MMP-14(lo) TIMP-3(hi) cells. We investigated the implications of these findings for human atherosclerosis. In vitro generated macrophages and foam-cell macrophages, together with atherosclerotic plaques characterised as unstable or stable, were examined for expression of MMP-14, TIMP-3, and inflammatory markers. Proinflammatory stimuli increased MMP-14 and decreased TIMP-3 mRNA and protein expression in human macrophages. However, conversion to foam-cells with oxidized LDL increased MMP-14 and decreased TIMP-3 protein, independently of inflammatory mediators and partly through posttranscriptional mechanisms. Within atherosclerotic plaques, MMP-14 was prominent in foam-cells with either pro- or anti-inflammatory macrophage markers, whereas TIMP-3 was present in less foamy macrophages and colocalised with CD206. MMP-14 positive macrophages were more abundant whereas TIMP-3 positive macrophages were less abundant in plaques histologically designated as rupture prone. We conclude that foam-cells characterised by high MMP-14 and low TIMP-3 expression are prevalent in rupture-prone atherosclerotic plaques, independent of pro- or anti-inflammatory activation. Therefore reducing MMP-14 activity and increasing that of TIMP-3 could be valid therapeutic approaches to reduce plaque rupture and myocardial infarction.


Asunto(s)
Macrófagos/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Placa Aterosclerótica/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Células Cultivadas , Humanos , Inmunohistoquímica , Activación de Macrófagos/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Circ Res ; 108(3): 284-93, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21164105

RESUMEN

RATIONALE: Homing of proangiogenic cells (PACs) is guided by chemoattractants and requires proteases to disrupt the extracellular matrix. The possibility that PAC recruitment involves an interaction between proteases and chemotactic factor receptors remains largely unexplored. OBJECTIVE: To determine the role of human tissue kallikrein (hK1) in PAC invasion and its dependency on kinin receptor signaling. METHODS AND RESULTS: Human mononuclear cells (MNCs) and culture-selected PACs express and release mature hK1 protein. HK1 gene (KLK1) silencing reduced PACs migratory, invasive, and proangiogenic activities. KLK1-knockout mouse bone marrow-derived MNCs showed similar impairments and were unable to support reparative angiogenesis in a mouse model of peripheral ischemia. Conversely, adenovirus-mediated KLK1 (Ad.KLK1) gene transfer enhanced PAC-associated functions, whereas the catalytically inactive variant R53H-KLK1 was ineffective. HK1-induced effects are mediated by a kinin B(2) receptor (B(2)R)-dependent mechanism involving inducible nitric oxide synthase and metalloproteinase-2 (MMP2). Lower hK1 protein levels were observed in PACs from type 2 diabetic (T2D) patients, whereas KLK1 mRNA levels were similar to those of healthy subjects, suggesting a post-transcriptional defect. Furthermore, B(2)R is normally expressed on T2D-PACs but remains uncoupled from downstream signaling. Importantly, whereas Ad.KLK1 alone could not restore T2D-PAC invasion capacity, combined KLK1 and B(2)R expression rescued the diabetic phenotype. CONCLUSIONS: This study reveals new interactive components of the PACs invasive machinery, acting via protease- and kinin receptor-dependent mechanisms.


Asunto(s)
Movimiento Celular/fisiología , Calicreínas/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Neovascularización Fisiológica/fisiología , Adulto , Anciano , Animales , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Regulación hacia Abajo , Femenino , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/metabolismo , Calicreínas/genética , Cininas/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Modelos Animales , Óxido Nítrico Sintasa/metabolismo , ARN Mensajero/metabolismo , Receptor de Bradiquinina B2/metabolismo , Transducción de Señal/fisiología
7.
Circulation ; 123(3): 282-91, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21220732

RESUMEN

BACKGROUND: Diabetes mellitus impairs endothelial cell (EC) function and postischemic reparative neovascularization by molecular mechanisms that are not fully understood. microRNAs negatively regulate the expression of target genes mainly by interaction in their 3' untranslated region. METHODS AND RESULTS: We found that microRNA-503 (miR-503) expression in ECs is upregulated in culture conditions mimicking diabetes mellitus (high D-glucose) and ischemia-associated starvation (low growth factors). Under normal culture conditions, lentivirus-mediated miR-503-forced expression inhibited EC proliferation, migration, and network formation on Matrigel (comparisons versus lentivirus.GFP control). Conversely, blocking miR-503 activity by either adenovirus-mediated transfer of a miR-503 decoy (Ad.decoymiR-503) or by antimiR-503 (antisense oligonucleotide) improved the functional capacities of ECs cultured under high D-glucose/low growth factors. We identified CCNE1 and cdc25A as direct miR-503 targets which are downregulated by high glucose/low growth factors in ECs. Next, we obtained evidence that miR-503 expression is increased in ischemic limb muscles of streptozotocin-diabetic mice and in ECs enriched from these muscles. Moreover, Ad.decoymiR-503 delivery to the ischemic adductor of diabetic mice corrected diabetes mellitus-induced impairment of postischemic angiogenesis and blood flow recovery. We finally investigated miR-503 and target gene expression in muscular specimens from the amputated ischemic legs of diabetic patients. As controls, calf biopsies of nondiabetic and nonischemic patients undergoing saphenous vein stripping were used. In diabetic muscles, miR-503 expression was remarkably higher, and it inversely correlated with cdc25 protein expression. Plasma miR-503 levels were also elevated in the diabetic individuals. CONCLUSIONS: Our data suggest miR-503 as a possible therapeutic target in diabetic patients with critical limb ischemia.


Asunto(s)
Diabetes Mellitus Experimental , Isquemia , MicroARNs/fisiología , Neovascularización Fisiológica/fisiología , Animales , Biopsia , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Células Endoteliales/citología , Células Endoteliales/fisiología , Extremidades/irrigación sanguínea , Expresión Génica/fisiología , Glucosa/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Isquemia/genética , Isquemia/patología , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Venas Umbilicales/citología , Fosfatasas cdc25/genética
8.
Biochem J ; 436(1): 133-43, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21352097

RESUMEN

Cell adhesion is essential for cell cycle progression in most normal cells. Loss of adhesion dependence is a hallmark of cellular transformation. The F-box protein Skp2 (S-phase kinase-associated protein 2) controls G(1)-S-phase progression and is subject to adhesion-dependent transcriptional regulation, although the mechanisms are poorly understood. We identify two cross-species conserved binding elements for the STAF (selenocysteine tRNA gene transcription-activating factor) in the Skp2 promoter that are essential for Skp2 promoter activity. Endogenous STAF specifically binds these elements in EMSA (electrophoretic mobility-shift assay) and ChIP (chromatin immunoprecipitation) analysis. STAF is sufficient and necessary for Skp2 promoter activity since exogenous STAF activates promoter activity and expression and STAF siRNA (small interfering RNA) inhibits Skp2 promoter activity, mRNA and protein expression and cell proliferation. Furthermore, ectopic Skp2 expression completely reverses the inhibitory effects of STAF silencing on proliferation. Importantly, STAF expression and binding to the Skp2 promoter is adhesion-dependent and associated with adhesion-dependent Skp2 expression in non-transformed cells. Ectopic STAF rescues Skp2 expression in suspension cells. Taken together, these results demonstrate that STAF is essential and sufficient for Skp2 promoter activity and plays a role in the adhesion-dependent expression of Skp2 and ultimately cell proliferation.


Asunto(s)
Proteínas Quinasas Asociadas a Fase-S/genética , Transactivadores/metabolismo , Transcripción Genética , Animales , Adhesión Celular , Proliferación Celular , Ensayo de Cambio de Movilidad Electroforética , Humanos , Masculino , Regiones Promotoras Genéticas , Ratas , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Transactivadores/genética , Transfección
9.
Biochim Biophys Acta Mol Cell Res ; 1869(11): 119329, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35905788

RESUMEN

Cardiac fibrosis is associated with increased stiffness of the myocardial extracellular matrix (ECM) in part mediated by increased cardiac fibroblast proliferation However, our understanding of the mechanisms regulating cardiac fibroblast proliferation are incomplete. Here we characterise a novel mechanism involving a combined activation of Yes-associated protein (YAP) targets RUNX Family Transcription Factor 2 (RUNX2) and TEA Domain Transcription Factor (TEAD). We demonstrate that cardiac fibroblast proliferation is enhanced by interaction with a stiff ECM compared to a soft ECM. This is associated with activation of the transcriptional co-factor, YAP. We demonstrate that this stiffness induced activation of YAP enhances the transcriptional activity of both TEAD and RUNX2 transcription factors. Inhibition of either TEAD or RUNX2, using gene silencing, expression of dominant-negative mutants or pharmacological inhibition, reduces cardiac fibroblast proliferation. Using mutants of YAP, defective in TEAD or RUNX2 activation ability, we demonstrate a dual role of YAP-mediated activation of TEAD and RUNX2 for substrate stiffness induced cardiac fibroblast proliferation. Our data highlights a previously unrecognised role of YAP mediated RUNX2 activation for cardiac fibroblast proliferation in response to increased ECM stiffness.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proliferación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Fibroblastos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP
10.
J Mol Cell Cardiol ; 50(1): 87-98, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20971121

RESUMEN

Cyclic AMP signalling promotes VSMC quiescence in healthy vessels and during vascular healing following injury. Cyclic AMP inhibits VSMC proliferation via mechanisms that are not fully understood. We investigated the role of PKA and Epac signalling on cAMP-induced inhibition of VSMC proliferation. cAMP-mediated growth arrest was PKA-dependent. However, selective PKA activation with 6-Benzoyl-cAMP did not inhibit VSMC proliferation, indicating a requirement for additional pathways. Epac activation using the selective cAMP analogue 8-CPT-2'-O-Me-cAMP, did not affect levels of hyperphosphorylated Retinoblastoma (Rb) protein, a marker of G1-S phase transition, or BrdU incorporation, despite activation of the Epac-effector Rap1. However, 6-Benzoyl-cAMP and 8-CPT-2'-O-Me-cAMP acted synergistically to inhibit Rb-hyperphosphorylation and BrdU incorporation, indicating that both pathways are required for growth inhibition. Consistent with this, constitutively active Epac increased Rap1 activity and synergised with 6-Benzoyl-cAMP to inhibit VSMC proliferation. PKA and Epac synergised to inhibit phosphorylation of ERK and JNK. Induction of stellate morphology, previously associated with cAMP-mediated growth arrest, was also dependent on activation of both PKA and Epac. Rap1 inhibition with Rap1GAP or siRNA silencing did not negate forskolin-induced inhibition of Rb-hyperphosphorylation, BrdU incorporation or stellate morphology. This data demonstrates for the first time that Epac synergises with PKA via a Rap1-independent mechanism to mediate cAMP-induced growth arrest in VSMC. This work highlights the role of Epac as a major player in cAMP-dependent growth arrest in VSMC.


Asunto(s)
Acetilcisteína/análogos & derivados , Proliferación Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Eritromicina/análogos & derivados , Miocitos del Músculo Liso/efectos de los fármacos , Acetilcisteína/metabolismo , Animales , Western Blotting , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Eritromicina/metabolismo , Masculino , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Paxillin/metabolismo , Faloidina/metabolismo , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/metabolismo
11.
Circ Res ; 103(2): e15-26, 2008 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-18566344

RESUMEN

Diabetes impairs endothelial function and reparative neovascularization. The p75 receptor of neurotrophins (p75(NTR)), which is scarcely present in healthy endothelial cells (ECs), becomes strongly expressed by capillary ECs after induction of peripheral ischemia in type-1 diabetic mice. Here, we show that gene transfer-induced p75(NTR) expression impairs the survival, proliferation, migration, and adhesion capacities of cultured ECs and endothelial progenitor cells (EPCs) and inhibits angiogenesis in vitro. Moreover, intramuscular p75(NTR) gene delivery impairs neovascularization and blood flow recovery in a mouse model of limb ischemia. These disturbed functions are associated with suppression of signaling mechanisms implicated in EC survival and angiogenesis. In fact, p75(NTR) depresses the VEGF-A/Akt/eNOS/NO pathway and additionally reduces the mRNA levels of ITGB1 [beta (1) integrin], BIRC5 (survivin), PTTG1 (securin) and VEZF1. Diabetic mice, which typically show impaired postischemic muscular neovascularization and blood perfusion recovery, have these defects corrected by intramuscular gene transfer of a dominant negative mutant form of p75(NTR). Collectively, our data newly demonstrate the antiangiogenic action of p75(NTR) and open new avenues for the therapeutic use of p75(NTR) inhibition to combat diabetes-induced microvascular liabilities.


Asunto(s)
Apoptosis/fisiología , Angiopatías Diabéticas/metabolismo , Endotelio Vascular/fisiología , Isquemia/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Receptor de Factor de Crecimiento Nervioso/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Humanos , Isquemia/etiología , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Factor de Crecimiento Nervioso/genética , Transducción de Señal/fisiología , Estreptozocina , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 29(2): 195-201, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19008530

RESUMEN

OBJECTIVE: Vascular smooth muscle cell (VSMC) apoptosis contributes to atherosclerotic plaque instability and myocardial infarction. Consequently, reducing VSMC apoptosis may be beneficial for reducing plaque instability and acute coronary events. We previously demonstrated that N-cadherin, a cell-cell adhesion molecule, reduces VSMC apoptosis in vitro. In this study, we examined whether a soluble form of N-cadherin (SNC) affected VSMC apoptosis and plaque stability. METHODS AND RESULTS: SNC significantly inhibited VSMC apoptosis in vitro by approximately 50% via activation of fibroblast growth factor receptor, phosphoinositide-3 kinase, and Akt signaling. SNC also significantly reduced macrophage and foam cell-macrophage apoptosis in vitro by >50%, without affecting monocyte invasion or macrophage proliferation. Elevation of plasma levels of SNC in male apolipoprotein E-deficient mice with existing atherosclerosis via adenoviral delivery significantly reduced VSMC and macrophage apoptosis in brachiocephalic artery plaques by approximately 60%. Additionally, SNC promoted plaques of a more stable phenotype by elevating VSMC:macrophage ratio and presence of VSMC-rich fibrous cap, as well as attenuating macrophage number and incidence of buried fibrous caps (a surrogate plaque rupture marker). CONCLUSIONS: In summary, this study demonstrates that SNC suppressed plaque instability by attenuation of apoptosis, suggesting that SNC may have a therapeutic potential for retarding plaque instability.


Asunto(s)
Apoptosis , Aterosclerosis/terapia , Tronco Braquiocefálico/patología , Cadherinas/biosíntesis , Terapia Genética/métodos , Adenoviridae/genética , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Tronco Braquiocefálico/metabolismo , Células CHO , Cadherinas/genética , Adhesión Celular , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Células Espumosas/metabolismo , Células Espumosas/patología , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/patología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fragmentos de Péptidos/biosíntesis , Transducción de Señal , Factores de Tiempo , Transducción Genética , Regulación hacia Arriba
13.
Arterioscler Thromb Vasc Biol ; 29(5): 657-64, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19164804

RESUMEN

OBJECTIVE: Human Tissue Kallikrein (hKLK1) overexpression promotes an enduring neovascularization of ischemic tissue, yet the cellular mechanisms of hKLK1-induced arteriogenesis remain unknown. Furthermore, no previous study has compared the angiogenic potency of hKLK1, with its loss of function polymorphic variant, rs5515 (R53H), which possesses reduced kinin-forming activity. METHODS AND RESULTS: Here, we demonstrate that tissue kallikrein knockout mice (KLK1-/-) show impaired muscle neovascularization in response to hindlimb ischemia. Gene-transfer of wild-type Ad.hKLK1 but not Ad.R53H-hKLK1 was able to rescue this defect. Similarly, in the rat mesenteric assay, Ad.hKLK1 induced a mature neovasculature with increased vessel diameter through kinin-B2 receptor-mediated recruitment of pericytes and vascular smooth muscle cells, whereas Ad.R53H-hKLK1 was ineffective. Moreover, hKLK1 but not R53H-hKLK1 overexpression in the zebrafish induced endothelial precursor cell migration and vascular remodeling. Furthermore, Ad.hKLK1 activates metalloproteinase (MMP) activity in normoperfused muscle and fails to promote reparative neovascularization in ischemic MMP9-/- mice, whereas its proarteriogenic action was preserved in ApoE-/- mice, an atherosclerotic model of impaired angiogenesis. CONCLUSIONS: These results demonstrate the fundamental role of endogenous Tissue Kallikrein in vascular repair and provide novel information on the cellular and molecular mechanisms responsible for the robust arterialization induced by hKLK1 overexpression.


Asunto(s)
Miembro Posterior/irrigación sanguínea , Neovascularización Fisiológica/fisiología , Circulación Esplácnica/fisiología , Calicreínas de Tejido/fisiología , Animales , Humanos , Isquemia/fisiopatología , Sistema Calicreína-Quinina/fisiología , Masculino , Metaloproteinasa 9 de la Matriz/fisiología , Ratones , Ratones Noqueados , Ratas , Cicatrización de Heridas/fisiología , Pez Cebra
14.
Biochim Biophys Acta Mol Cell Res ; 1867(7): 118691, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32119877

RESUMEN

Actin dynamics regulate cell behaviour in response to physiological signals. Here we demonstrate a novel role for nuclear actin in inhibiting cell proliferation and migration. We demonstrate that physiological signals that elevate cAMP, which is anti-mitogenic in vascular smooth muscle cells, increases nuclear actin monomer levels. Expression of a nuclear-targeted polymerisation-defective actin mutant (NLS-ActinR62D) inhibited proliferation and migration. Preventing nuclear actin monomer accumulation by enhancing its nuclear export or polymerisation reversed the anti-mitogenic and anti-migratory effects of cAMP. Transcriptomic analysis identified repression of proliferation and migration associated genes regulated by serum response factor (SRF) and TEA Domain (TEAD) transcription factors. Accordingly, NLS-ActinR62D inhibited SRF and TEAD activity and target gene expression, and these effects were reversed by constitutively-active mutants of the TEAD and SRF co-factors YAP, TAZ and MKL1. In summary, intranuclear actin inhibits proliferation and migration by inhibiting YAP-TEAD and MKL-SRF activity. This mechanism explains the anti-mitogenic and anti-migratory properties of physiological signals that elevate cAMP. SUMMARY: McNeill et al show that increased levels of intranuclear actin monomer inhibit cell proliferation and migration by inhibiting MKL1-SRF and YAP/TAZ-TEAD-dependent gene expression. This mechanism mediates the anti-mitogenic and anti-migratory effects of physiological signals that elevate cyclic-AMP.


Asunto(s)
Actinas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Unión al ADN/genética , Proteínas Nucleares/genética , Factor de Respuesta Sérica/genética , Factores de Transcripción/genética , Movimiento Celular/genética , Núcleo Celular/genética , Proliferación Celular/genética , AMP Cíclico/genética , Regulación de la Expresión Génica/genética , Humanos , Factores de Transcripción de Dominio TEA , Proteínas Señalizadoras YAP
15.
Thromb Haemost ; 101(6): 1006-11, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19492140

RESUMEN

Plaque rupture underlies most myocardial infarctions. Plaques vulnerable to rupture have thin fibrous caps, an excess of macrophages over vascular smooth muscle cells, large lipid cores, and depletion of collagen and other matrix proteins form the cap and lipid core. Production of matrix metalloproteinases from macrophages is prominent in human plaques, and studies in genetically modified mice imply a causative role for metalloproteinases in plaque vulnerability. Recent in-vitro studies on human monocyte-derived macrophages and on foam-cell macrophages generated in vivo suggest the existence of several macrophage phenotypes with distinct patterns of metalloproteinase expression. These phenotypes could play differing roles in cap, core and aneurysm formation.


Asunto(s)
Aterosclerosis/enzimología , Aterosclerosis/patología , Células Espumosas/enzimología , Metaloproteinasas de la Matriz/metabolismo , Animales , Rotura de la Aorta/etiología , Rotura de la Aorta/prevención & control , Aterosclerosis/complicaciones , Aterosclerosis/inmunología , Células Espumosas/patología , Regulación Enzimológica de la Expresión Génica , Humanos , Metabolismo de los Lípidos , Metaloproteinasas de la Matriz/genética , Ratones , Modelos Inmunológicos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/enzimología , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Miocitos del Músculo Liso/patología , Inhibidores Tisulares de Metaloproteinasas/inmunología , Inhibidores Tisulares de Metaloproteinasas/metabolismo
16.
J Vasc Surg ; 50(5): 1135-42, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19878790

RESUMEN

OBJECTIVE: Vascular smooth muscle cell (VSMC) proliferation plays an important role in the development of postangioplasty or in-stent restenosis, venous graft failure, and atherosclerosis. Our previous work has demonstrated S-phase kinase-associated protein-2 (Skp2), an F-box subunit of SCF(Skp2) ubiquitin ligase, as an important mediator and common final pathway for growth factors, extracellular matrices, and cyclic-nucleotides to regulate VSMC proliferation in vitro. However, whether alteration of Skp2 function also regulates VSMC proliferation in vivo and neointimal thickening postvascular injury remains unclear. We investigated the effect of Skp2 on VSMC proliferation and neointimal formation in vivo. METHODS AND RESULTS: Firstly, we demonstrated that Skp2-null mice developed significantly smaller neointimal areas than wild-type mice after carotid ligation. Secondly, to further identify a local rather than a systemic effect of Skp2 alteration, we demonstrated that adenovirus-mediated expression of dominant-negative Skp2 in the balloon-injured rat carotid artery significantly increased medial p27(Kip1) levels, inhibited VSMC proliferation, and the subsequent neointimal thickening. Lastly, to determine if Skp2 alone is sufficient to drive VSMC proliferation and lesion development in vivo, we demonstrated that adenovirus-delivery of wild-type Skp2 to the minimally-injured rat carotids is sufficient to downregulate p27(Kip1) protein levels, enhanced medial VSMC proliferation, and the neointimal thickening. CONCLUSION: This data provides, we believe for the first time, a more comprehensive understanding of Skp2 in the regulation of VSMC proliferation and neointimal formation and suggests that Skp2 is a promising target in the treatment of vasculoproliferative diseases.


Asunto(s)
Traumatismos de las Arterias Carótidas/metabolismo , Proliferación Celular , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Túnica Íntima/metabolismo , Adenoviridae/genética , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Vectores Genéticos , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas Asociadas a Fase-S/genética , Transducción Genética , Túnica Íntima/patología
17.
Arterioscler Thromb Vasc Biol ; 28(9): 1647-53, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18566294

RESUMEN

OBJECTIVE: An excess of metalloproteinases (MMPs) over tissue inhibitors of metalloproteinases (TIMPs) may favor atherosclerotic plaque rupture. We compared TIMP levels in nonfoamy and foam-cell macrophages (FCM) generated in vivo. METHODS AND RESULTS: In vivo generated rabbit FCM exhibited 84% reduced TIMP-3 protein compared to nonfoamy macrophages, and immunocytochemistry revealed a TIMP-3 negative subset (28%). Strikingly, only TIMP-3 negative FCM invaded a synthetic basement membrane, and invasion was inhibited by exogenous TIMP-3. TIMP-3 negative FCM also had increased proliferation and apoptosis rates compared to TIMP-3 positive cells, which were retarded by exogenous TIMP-3; this also reduced gelatinolytic activity. TIMP-3 negative FCM were found at the base of advanced rabbit plaques and in the rupture-prone shoulders of human plaques. To explain the actions of low TIMP-3 we observed a 26-fold increase in MT1-MMP (MMP-14) protein in FCM. Adding an MT1-MMP neutralizing antibody reduced foam-cell invasion, apoptosis, and gelatinolytic activity. Furthermore, MT1-MMP overexpressing and TIMP-3 negative FCM were found at the same locations in atherosclerotic plaques. CONCLUSIONS: These results demonstrate that TIMP-3 is downregulated in a distinct subpopulation of FCM which have increased MMP-14. These cells are highly invasive and have increased proliferation and apoptosis, all properties expected to destabilise atherosclerotic plaques.


Asunto(s)
Aterosclerosis/enzimología , Movimiento Celular , Células Espumosas/enzimología , Metaloproteinasa 14 de la Matriz/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Animales , Apoptosis , Aterosclerosis/patología , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo , Células Espumosas/patología , Humanos , Inmunohistoquímica , ARN Mensajero/metabolismo , Conejos , Inhibidor Tisular de Metaloproteinasa-3/genética , Regulación hacia Arriba
18.
Cardiovasc Res ; 80(2): 290-8, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18599477

RESUMEN

AIMS: Vascular smooth muscle cell (VSMC) proliferation contributes to intima formation after angioplasty or venous by-pass grafting, and during atherosclerosis. VSMC proliferation requires degradation of p27(Kip1) promoted by S-phase kinase-associated protein-2 (Skp2), an F-box protein component of the Skp-Cullin-F-box(Skp2) ubiquitin-ligase. We investigated the role of Rac(1) in the regulation of Skp2 in rat VSMC. METHODS AND RESULTS: Rat carotid balloon injury increased Rac(1) activity. Rho GTPase inhibition with Clostridium difficile Toxin B or specific Rac(1) inhibition with adenovirus-mediated expression of dominant-negative Rac(1) reduced Skp2 levels, and VSMC proliferation in vitro and intima formation in vivo following carotid balloon injury. Inhibition of Skp2 expression and proliferation by dominant-negative Rac(1) was reversed by exogenous Skp2. Elevation of endogenous adenosine 3',5'-cyclic monophosphate (cAMP) with forskolin-inhibited Rac(1) activity, reduced Skp2, increased p27(Kip1) and inhibited VSMC proliferation, effects that were reversed by constitutively active Rac(1). These effects were independent of Rac(1) Cdc42/Rac interactive binding (CRIB)-domain effector proteins but associated with Rac(1)-dependent actin polymerization. CONCLUSION: Rac(1) activity regulates VSMC proliferation by controlling Skp2 levels. Activation of Rac(1) induced by balloon injury in vivo increases Skp2 levels, which promotes VSMC proliferation and intima formation. Inhibition of this novel pathway underlies the negative effects of cAMP on VSMC proliferation.


Asunto(s)
Traumatismos de las Arterias Carótidas/enzimología , Arteria Carótida Común/enzimología , Proliferación Celular , Músculo Liso Vascular/enzimología , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Túnica Íntima/enzimología , Proteína de Unión al GTP rac1/metabolismo , Actinas/metabolismo , Animales , Proteínas Bacterianas/farmacología , Toxinas Bacterianas/farmacología , Traumatismos de las Arterias Carótidas/etiología , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/efectos de los fármacos , Arteria Carótida Común/patología , Cateterismo/efectos adversos , Células Cultivadas , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Mutación , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas Asociadas a Fase-S/genética , Transducción de Señal , Factores de Tiempo , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética
19.
Circ Res ; 98(9): 1141-50, 2006 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-16574903

RESUMEN

Cyclic nucleotides inhibit vascular smooth muscle cell (VSMC) proliferation but the underlying molecular mechanisms are incompletely understood. We studied the role of S-phase kinase-associated protein-2 (Skp2), an F-box protein of SCFSkp2 ubiquitin ligase responsible for polyubiquitylation of and subsequent proteolysis of p27Kip1, a key step leading to cell cycle progression. Skp2 mRNA and protein were upregulated in mitogen-stimulated VSMCs and after balloon injury in rat carotid arteries, where the time course and location of Skp2 expression closely paralleled that of proliferating cell nuclear antigen. Skp2 small interference RNA (siRNA) reduced Skp2 expression, increased p27Kip1 levels, and inhibited VSMC proliferation in vitro. cAMP-elevating agents prominently inhibited VSMC proliferation and Skp2 expression through inhibiting Skp2 transcription as well as decreasing Skp2 protein stability. Consistent with this, activation of protein kinase A, a downstream target of cAMP, was shown to negatively regulate focal adhesion kinase (FAK) phosphorylation and Skp2 expression. Adenovirus-mediated Skp2 expression reversed cAMP-induced p27Kip1 upregulation and rescued cAMP-related S-phase entry inhibition up to 50%. 8-bromo-cGMP also moderately reduced Skp2 and cell proliferation when VSMCs were incubated with low serum concentration. Interestingly, we showed that 8-bromo-cGMP inhibited Skp2 expression also through activation of protein kinase A, not protein kinase G, which conversely enhanced FAKY397 phosphorylation and Skp2 expression. After balloon injury of rat carotid arteries, local forskolin treatment significantly reduced FAKY397 phosphorylation, Skp2 expression, VSMC proliferation, and subsequent neointimal thickening. These data demonstrate for the first time that Skp2 is an important factor in VSMC proliferation and its inhibition by cyclic nucleotides.


Asunto(s)
Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/patología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Nucleótidos Cíclicos/farmacología , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Animales , Sangre , Bucladesina/farmacología , Cateterismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Estabilidad de Medicamentos , Masculino , Músculo Liso Vascular/enzimología , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas Asociadas a Fase-S/química , Proteínas Quinasas Asociadas a Fase-S/genética , Transcripción Genética/efectos de los fármacos , Túnica Íntima/patología , Regulación hacia Arriba
20.
Circ Res ; 99(12): 1329-37, 2006 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-17122440

RESUMEN

We previously observed that stimulation of vascular smooth muscle cell (VSMC) proliferation with growth factors is associated with dismantling of cadherin junctions and nuclear translocation of beta-catenin. In this study we demonstrate directly that growth factors stimulate beta-catenin/T-cell factor (TCF) signaling in primary VSMCs. To determine whether beta-catenin/TCF signaling regulates VSMC proliferation via modulation of the beta-catenin/TCF responsive cell cycle genes, cyclin D1 and p21, we inhibited beta-catenin/TCF signaling by adenoviral-mediated over-expression of N-Cadherin, ICAT (an endogenous inhibitor of beta-catenin/TCF signaling), or a dominant negative (dn) mutant of TCF-4. N-cadherin, ICAT or dnTCF-4 over-expression significantly reduced proliferation of isolated human VSMCs by approximately 55%, 80%, and 45% respectively. Similar effects were observed in human saphenous vein medial segments where proliferation was reduced by approximately 55%. Transfection of dnTCF-4 in the ISS10 human VSMC line significantly lowered TCF and cyclin D1 reporter activity but significantly elevated p21 reporter activity, indicating regulation of these genes by beta-catenin/TCF signaling. In support of this, over-expression of N-cadherin, ICAT or dnTCF-4 in isolated human VSMCs significantly lowered levels of cyclin D1 mRNA and protein levels. In contrast, over-expression of N-Cadherin, ICAT or dnTCF4 significantly elevated p21 mRNA and protein levels. In summary, we have demonstrated that increasing N-cadherin and inhibiting beta-catenin/TCF signaling reduces VSMC proliferation, decreases the expression of cyclin D1 and increases levels of the cell cycle inhibitor, p21. We therefore suggest that the N-cadherin and beta-catenin/TCF signaling pathway is a key modulator of VSMC proliferation via regulation of these 2 beta-catenin/TCF responsive genes.


Asunto(s)
Ciclina D1/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Músculo Liso Vascular/fisiología , Factores de Transcripción TCF/metabolismo , beta Catenina/metabolismo , Aorta/citología , División Celular/fisiología , Células Cultivadas , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Músculo Liso Vascular/citología , Vena Safena/citología , Transducción de Señal/fisiología , Transcripción Genética/fisiología , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología , Enfermedades Vasculares/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA