Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
2.
Nature ; 495(7439): 107-10, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23426265

RESUMEN

Ageing is the predominant risk factor for cardiovascular diseases and contributes to a significantly worse outcome in patients with acute myocardial infarction. MicroRNAs (miRNAs) have emerged as crucial regulators of cardiovascular function and some miRNAs have key roles in ageing. We propose that altered expression of miRNAs in the heart during ageing contributes to the age-dependent decline in cardiac function. Here we show that miR-34a is induced in the ageing heart and that in vivo silencing or genetic deletion of miR-34a reduces age-associated cardiomyocyte cell death. Moreover, miR-34a inhibition reduces cell death and fibrosis following acute myocardial infarction and improves recovery of myocardial function. Mechanistically, we identified PNUTS (also known as PPP1R10) as a novel direct miR-34a target, which reduces telomere shortening, DNA damage responses and cardiomyocyte apoptosis, and improves functional recovery after acute myocardial infarction. Together, these results identify age-induced expression of miR-34a and inhibition of its target PNUTS as a key mechanism that regulates cardiac contractile function during ageing and after acute myocardial infarction, by inducing DNA damage responses and telomere attrition.


Asunto(s)
Envejecimiento/fisiología , Regulación de la Expresión Génica , Corazón/fisiología , MicroARNs/genética , Miocardio/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Animales , Apoptosis , Daño del ADN , Fibrosis/genética , Fibrosis/patología , Eliminación de Gen , Técnicas de Inactivación de Genes , Terapia Genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Especificidad por Sustrato , Telómero/genética , Telómero/metabolismo
3.
Basic Res Cardiol ; 114(1): 1, 2018 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-30443679

RESUMEN

Monocytes are involved in adverse left ventricular (LV) remodelling following myocardial infarction (MI). To provide therapeutic opportunities we aimed to identify gene transcripts in monocytes that relate to post-MI healing and evaluated intervention with the observed gene activity in a rat MI model. In 51 MI patients treated by primary percutaneous coronary intervention (PCI), the change in LV end-diastolic volume index (EDVi) from baseline to 4-month follow-up was assessed using cardiovascular magnetic resonance imaging (CMR). Circulating monocytes were collected at day 5 (Arterioscler Thromb Vasc Biol 35:1066-1070, 2015; Cell Stem Cell 16:477-487, 2015; Curr Med Chem 13:1877-1893, 2006) after primary PCI for transcriptome analysis. Transcriptional profiling and pathway analysis revealed that patients with a decreased LV EDVi showed an induction of type I interferon (IFN) signalling (type I IFN pathway: P value < 0.001; false discovery rate < 0.001). We subsequently administered 15,000 Units of IFN-α subcutaneously in a rat MI model for three consecutive days following MI. Cardiac function was measured using echocardiography and infarct size/cardiac inflammation using (immuno)-histochemical analysis. We found that IFN-α application deteriorated ventricular dilatation and increased infarct size at day 28 post-MI. Moreover, IFN-α changed the peripheral monocyte subset distribution towards the pro-inflammatory monocyte subset whereas in the myocardium, the presence of the alternative macrophage subset was increased at day 3 post-MI. Our findings suggest that induction of type I IFN signalling in human monocytes coincides with adverse LV remodelling. In rats, however, IFN-α administration deteriorated post-MI healing. These findings underscore important but also contradictory roles for the type I IFN response during cardiac healing following MI.


Asunto(s)
Interferón Tipo I/metabolismo , Monocitos/trasplante , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Remodelación Ventricular , Adulto , Anciano , Animales , Trasplante de Médula Ósea/métodos , Femenino , Humanos , Interferón Tipo I/farmacología , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Infarto del Miocardio/patología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Remodelación Ventricular/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
4.
Am J Respir Crit Care Med ; 193(12): 1410-20, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-26760925

RESUMEN

RATIONALE: Altered pulmonary hemodynamics and fluid flow-induced high shear stress (HSS) are characteristic hallmarks in the pathogenesis of pulmonary arterial hypertension (PAH). However, the contribution of HSS to cellular and vascular alterations in PAH is unclear. OBJECTIVES: We hypothesize that failing shear adaptation is an essential part of the endothelial dysfunction in all forms of PAH and tested whether microvascular endothelial cells (MVECs) or pulmonary arterial endothelial cells (PAECs) from lungs of patients with PAH adapt to HSS and if the shear defect partakes in vascular remodeling in vivo. METHODS: PAH MVEC (n = 7) and PAH PAEC (n = 3) morphology, function, protein, and gene expressions were compared with control MVEC (n = 8) under static culture conditions and after 24, 72, and 120 hours of HSS. MEASUREMENTS AND MAIN RESULTS: PAH MVEC showed a significantly delayed morphological shear adaptation (P = 0.03) and evidence of cell injury at sites of nonuniform shear profiles that are critical loci for vascular remodeling in PAH. In clear contrast, PAEC isolated from the same PAH lungs showed no impairments. PAH MVEC gene expression and transcriptional shear activation were not altered but showed significant decreased protein levels (P = 0.02) and disturbed interendothelial localization of the shear sensor platelet endothelial cell adhesion molecule-1 (PECAM-1). The decreased PECAM-1 levels were caused by caspase-mediated cytoplasmic cleavage but not increased cell apoptosis. Caspase blockade stabilized PECAM-1 levels, restored endothelial shear responsiveness in vitro, and attenuated occlusive vascular remodeling in chronically hypoxic Sugen5416-treated rats modeling severe PAH. CONCLUSIONS: Delayed shear adaptation, which promotes shear-induced endothelial injury, is a newly identified dysfunction specific to the microvascular endothelium in PAH. The shear response is normalized on stabilization of PECAM-1, which reverses intimal remodeling in vivo.


Asunto(s)
Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Microvasos/fisiopatología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Remodelación Vascular/fisiología , Adulto , Animales , Western Blotting , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Microvasos/metabolismo , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Ratas , Adulto Joven
5.
Basic Res Cardiol ; 111(4): 38, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27146510

RESUMEN

Coronary artery disease (CAD), also known as ischemic heart disease (IHD), is the leading cause of mortality in the western world, with developing countries showing a similar trend. With the increased understanding of the role of the immune system and inflammation in coronary artery disease, it was shown that macrophages play a major role in this disease. Costimulatory molecules are important regulators of inflammation, and especially, the CD40L-CD40 axis is of importance in the pathogenesis of cardiovascular disease. Although it was shown that CD40 can mediate macrophage function, its exact role in macrophage biology has not gained much attention in cardiovascular disease. Therefore, the goal of this review is to give an overview on the role of macrophage-specific CD40 in cardiovascular disease, with a focus on coronary artery disease. We will discuss the function of CD40 on the macrophage and its (proposed) role in the reduction of atherosclerosis, the reduction of neointima formation, and the stimulation of arteriogenesis.


Asunto(s)
Antígenos CD40/inmunología , Enfermedad de la Arteria Coronaria/patología , Macrófagos/patología , Animales , Enfermedad de la Arteria Coronaria/inmunología , Humanos , Macrófagos/inmunología
6.
Am J Physiol Heart Circ Physiol ; 309(10): H1667-78, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26432845

RESUMEN

Circulating angiogenic cells (CACs) are monocyte-derived cells with endothelial characteristics, which contribute to both angiogenesis and arteriogenesis in a paracrine way. Interferon-ß (IFN-ß) is known to inhibit these divergent processes in animals and patients. We hypothesized that IFN-ß might act by affecting the differentiation and function of CACs. CACs were cultured from peripheral blood mononuclear cells and phenotypically characterized by surface expression of monocytic and endothelial markers. IFN-ß significantly reduced the number of CACs by 18-64%. Apoptosis was not induced by IFN-ß, neither in mononuclear cells during differentiation, nor after maturation to CACs. Rather, IFN-ß impaired adhesion to, and spreading on, fibronectin, which was dependent on α5ß1 (VLA-5)-integrin. IFN-ß affected the function of VLA-5 in mature CACs, leading to rounding and detachment of cells, by induction of calpain 1 activity. Cell rounding and detachment was completely reversed by inhibition of calpain 1 activity in mature CACs. During in vitro capillary formation, CAC addition and calpain 1 inhibition enhanced sprouting of endothelial cells to a comparable extent, but were not sufficient to rescue tube formation in the presence of IFN-ß. We show that the IFN-ß-induced reduction of the numbers of in vitro differentiated CACs is based on activation of calpain 1, resulting in an attenuated adhesion to extracellular matrix proteins via VLA-5. In vivo, this could lead to inhibition of vessel formation due to reduction of the locally recruited CAC numbers and their paracrine angiogenic factors.


Asunto(s)
Calpaína/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Interferón beta/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Apoptosis/efectos de los fármacos , Calpaína/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Fibronectinas , Humanos , Técnicas In Vitro , Integrina alfa5beta1/efectos de los fármacos , Integrina alfa5beta1/metabolismo , Leucocitos Mononucleares/metabolismo , Neovascularización Fisiológica/fisiología
7.
Eur Heart J ; 35(6): 376-85, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23966310

RESUMEN

AIMS: Monocytes are critical mediators of healing following acute myocardial infarction (AMI), making them an interesting target to improve myocardial repair. The purpose of this study was a gain of insight into the source and recruitment of monocytes following AMI in humans. METHODS AND RESULTS: Post-mortem tissue specimens of myocardium, spleen and bone marrow were collected from 28 patients who died at different time points after AMI. Twelve patients who died from other causes served as controls. The presence and localization of monocytes (CD14(+) cells), and their CD14(+)CD16(-) and CD14(+)CD16(+) subsets, were evaluated by immunohistochemical and immunofluorescence analyses. CD14(+) cells localized at distinct regions of the infarcted myocardium in different phases of healing following AMI. In the inflammatory phase after AMI, CD14(+) cells were predominantly located in the infarct border zone, adjacent to cardiomyocytes, and consisted for 85% (78-92%) of CD14(+)CD16(-) cells. In contrast, in the subsequent post-AMI proliferative phase, massive accumulation of CD14(+) cells was observed in the infarct core, containing comparable proportions of both the CD14(+)CD16(-) [60% (31-67%)] and CD14(+)CD16(+) subsets [40% (33-69%)]. Importantly, in AMI patients, of the number of CD14(+) cells was decreased by 39% in the bone marrow and by 58% in the spleen, in comparison with control patients (P = 0.02 and <0.001, respectively). CONCLUSIONS: Overall, this study showed a unique spatiotemporal pattern of monocyte accumulation in the human myocardium following AMI that coincides with a marked depletion of monocytes from the spleen, suggesting that the human spleen contains an important reservoir function for monocytes.


Asunto(s)
Monocitos/fisiología , Infarto del Miocardio/patología , Bazo/fisiología , Anciano , Antígenos CD/metabolismo , Células de la Médula Ósea/fisiología , Estudios de Casos y Controles , Femenino , Humanos , Inmunohistoquímica , Masculino , Monocitos/clasificación , Infarto del Miocardio/inmunología , Miocardio/patología , Bazo/inmunología
8.
J Neurosci ; 33(16): 6857-63, 2013 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23595744

RESUMEN

Blood-brain barrier (BBB) dysfunction is a major hallmark of many neurological diseases, including multiple sclerosis (MS). Using a genomics approach, we defined a microRNA signature that is diminished at the BBB of MS patients. In particular, miR-125a-5p is a key regulator of brain endothelial tightness and immune cell efflux. Our findings suggest that repair of a disturbed BBB through microRNAs may represent a novel avenue for effective treatment of MS.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Encéfalo/patología , Células Endoteliales/fisiología , Inflamación/patología , MicroARNs/metabolismo , Esclerosis Múltiple/patología , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular Transformada , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/fisiología , Humanos , MicroARNs/genética , ARN Interferente Pequeño/farmacología , Migración Transendotelial y Transepitelial/efectos de los fármacos , Transfección
9.
J Mol Cell Cardiol ; 67: 94-102, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24389343

RESUMEN

Acute myocardial infarction (AMI) is accompanied by increased expression of Toll like receptors (TLR)-2 and TLR4 on circulating monocytes. In animal models, blocking TLR2/4 signaling reduces inflammatory cell influx and infarct size. The clinical consequences of TLR activation during AMI in humans are unknown, including its role in long-term cardiac functional outcome Therefore, we analyzed gene expression in whole blood samples from 28 patients with an acute ST elevation myocardial infarction (STEMI), enrolled in the EXenatide trial for AMI patients (EXAMI), both at admission and after 4-month follow-up, by whole genome expression profiling and real-time PCR. Cardiac function was determined by cardiac magnetic resonance (CMR) imaging at baseline and after 4-month follow-up. TLR pathway activation was shown by increased expression of TLR4 and its downstream genes, including IL-18R1, IL-18R2, IL-8, MMP9, HIF1A, and NFKBIA. In contrast, expression of the classical TLR-induced genes, TNF, was reduced. Bioinformatics analysis and in vitro experiments explained this noncanonical TLR response by identification of a pivotal role for HIF-1α. The extent of TLR activation and IL-18R1/2 expression in circulating cells preceded massive troponin-T release and correlated with the CMR-measured ischemic area (R=0.48, p=0.01). In conclusion, we identified a novel HIF-1-dependent noncanonical TLR activation pathway in circulating leukocytes leading to enhanced IL-18R expression which correlated with the magnitude of the ischemic area. This knowledge may contribute to our mechanistic understanding of the involvement of the innate immune system during STEMI and may yield diagnostic and prognostic value for patients with myocardial infarction.


Asunto(s)
Interleucina-18/metabolismo , Infarto del Miocardio/fisiopatología , Receptor Toll-Like 4/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Interleucina-18/sangre , Interleucina-18/genética , Leucocitos/metabolismo , Persona de Mediana Edad , Receptor Toll-Like 4/sangre , Receptor Toll-Like 4/genética , Regulación hacia Arriba
10.
Cardiovasc Diabetol ; 13: 150, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25361524

RESUMEN

BACKGROUND: Peroxisome proliferator-activated receptor-γ (PPARγ) agonists, which have been used as insulin sensitizers in diabetic patients, may improve functions of endothelial cells (ECs). We investigated the effect of PPARγ on angiogenic activities of murine ECs and bone marrow-derived proangiogenic cells (PACs). METHODS: PACs were isolated from bone marrow of 10-12 weeks old, wild type, db/db and PPARγ heterozygous animals. Cells were cultured on fibronectin and gelatin coated dishes in EGM-2MV medium. For in vitro stimulations, rosiglitazone (10 µmol/L) or GW9662 (10 µmol/L) were added to 80% confluent cell cultures for 24 hours. Angiogenic potential of PACs and ECs was tested in vitro and in vivo in wound healing assay and hind limb ischemia model. RESULTS: ECs and PACs isolated from diabetic db/db mice displayed a reduced angiogenic potential in ex vivo and in vitro assays, the effect partially rescued by incubation of cells with rosiglitazone (PPARγ activator). Correction of diabetes by administration of rosiglitazone in vivo did not improve angiogenic potential of isolated PACs or ECs. In a hind limb ischemia model we demonstrated that local injection of conditioned media harvested from wild type PACs improved the blood flow restoration in db/db mice, confirming the importance of paracrine action of the bone marrow-derived cells. CONCLUSIONS: In summary, activation of PPARγ by rosiglitazone improves angiogenic potential of diabetic ECs and PACs, but decreased expression of PPARγ in diabetes does not impair angiogenesis.


Asunto(s)
Células de la Médula Ósea/citología , Médula Ósea/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , PPAR gamma/metabolismo , Células Madre/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células Cultivadas , Células Endoteliales/metabolismo , Hipoglucemiantes/farmacología , Isquemia/tratamiento farmacológico , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , PPAR gamma/genética , Rosiglitazona , Células Madre/citología , Células Madre/efectos de los fármacos , Tiazolidinedionas/farmacología , Cicatrización de Heridas/efectos de los fármacos
11.
Eur Heart J ; 34(30): 2346-53, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23594591

RESUMEN

AIMS: Lack of gadolinium-contrast wash-in on first-pass perfusion imaging, early gadolinium-enhanced imaging, or late gadolinium-enhanced (LGE) cardiovascular magnetic resonance (CMR) imaging after revascularized ST-elevation myocardial infarction (STEMI) is commonly referred to as microvascular obstruction (MVO). Additionally, T2-weighted imaging allows for the visualization of infarct-related oedema and intramyocardial haemorrhage (IMH) within the infarction. However, the exact histopathological correlate of the contrast-devoid core and its relation to IMH is unknown. METHODS AND RESULTS: In eight Yorkshire swine, the circumflex coronary artery was occluded for 75 min by a balloon catheter. After 7 days, CMR with cine imaging, T2-weighted turbospinecho, and LGE was performed. Cardiovascular magnetic resonance images were compared with histological findings after phosphotungstic acid-haematoxylin and anti-CD31/haematoxylin staining. These findings were compared with CMR findings in 27 consecutive PCI-treated STEMI patients, using the same scanning protocol. In the porcine model, the infarct core contained extensive necrosis and erythrocyte extravasation, without intact vasculature and hence, no MVO. The surrounding-gadolinium-enhanced-area contained granulation tissue, leucocyte infiltration, and necrosis with morphological intact microvessels containing microthrombi, without erythrocyte extravasation. Areas with IMH (median size 1.92 [0.36-5.25] cm(3)) and MVO (median size 2.19 [0.40-4.58] cm(3)) showed close anatomic correlation [intraclass correlation coefficient (ICC) 0.85, r = 0.85, P = 0.03]. Of the 27 STEMI patients, 15 had IMH (median size 6.60 [2.49-9.79] cm(3)) and 16 had MVO (median size 4.31 [1.05-7.57] cm(3)). Again, IMH and MVO showed close anatomic correlation (ICC 0.87, r = 0.93, P < 0.001). CONCLUSION: The contrast-devoid core of revascularized STEMI contains extensive erythrocyte extravasation with microvascular damage. Attenuating the reperfusion-induced haemorrhage may be a novel target in future adjunctive STEMI treatment.


Asunto(s)
Cardiomiopatías/patología , Oclusión Coronaria/patología , Hemorragia/patología , Infarto del Miocardio/patología , Adulto , Anciano , Animales , Oclusión con Balón , Medios de Contraste , Trombosis Coronaria/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Angiografía por Resonancia Magnética , Imagen por Resonancia Cinemagnética , Masculino , Meglumina , Microvasos/patología , Persona de Mediana Edad , Infarto del Miocardio/terapia , Revascularización Miocárdica/efectos adversos , Necrosis/patología , Compuestos Organometálicos , Intervención Coronaria Percutánea , Sus scrofa
12.
Eur J Clin Invest ; 43(1): 100-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23083351

RESUMEN

BACKGROUND: The role of bone marrow-derived cells in stimulating angiogenesis, vascular repair or remodelling has been well established, but the nature of the circulating angiogenic cells is still controversial. DESIGN: The existing literature on different cell types that contribute to angiogenesis in multiple pathologies, most notably ischaemic and tumour angiogenesis, is reviewed, with a focus on subtypes of angiogenic mononuclear cells and their local recruitment and activation. RESULTS: A large number of different cells of myeloid origin support angiogenesis without incorporating permanently into the newly formed vessel, which distinguishes these circulating angiogenic cells (CAC) from endothelial progenitor cells (EPC). Although CAC frequently express individual endothelial markers, they all share multiple characteristics of monocytes and only express a limited set of discriminative surface markers in the circulation. When cultured ex vivo, or surrounding the angiogenic vessel in vivo, however, many of them acquire similar additional markers, making their discrimination in situ difficult. CONCLUSION: Different subsets of monocytes show angiogenic properties, but the distinct microenvironment, in vitro or in vivo, is needed for the development of their pro-angiogenic function.


Asunto(s)
Células Endoteliales/fisiología , Monocitos/fisiología , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Células Madre/fisiología , Humanos , Isquemia/fisiopatología
13.
Circ Res ; 109(10): 1115-9, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21903938

RESUMEN

RATIONALE: Aging represents a major risk factor for coronary artery disease and aortic aneurysm formation. MicroRNAs (miRs) have emerged as key regulators of biological processes, but their role in age-associated vascular pathologies is unknown. OBJECTIVE: We aim to identify miRs in the vasculature that are regulated by age and play a role in age-induced vascular pathologies. METHODS AND RESULTS: Expression profiling of aortic tissue of young versus old mice identified several age-associated miRs. Among the significantly regulated miRs, the increased expression of miR-29 family members was associated with a profound downregulation of numerous extracellular matrix (ECM) components in aortas of aged mice, suggesting that this miR family contributes to ECM loss, thereby sensitizing the aorta for aneurysm formation. Indeed, miR-29 expression was significantly induced in 2 experimental models for aortic dilation: angiotensin II-treated aged mice and genetically induced aneurysms in Fibulin-4(R/R) mice. More importantly, miR-29b levels were profoundly increased in biopsies of human thoracic aneurysms, obtained from patients with either bicuspid (n=79) or tricuspid aortic valves (n=30). Finally, LNA-modified antisense oligonucleotide-mediated silencing of miR-29 induced ECM expression and inhibited angiotensin II-induced dilation of the aorta in mice. CONCLUSION: In conclusion, miR-29-mediated downregulation of ECM proteins may sensitize the aorta to the formation of aneurysms in advanced age. Inhibition of miR-29 in vivo abrogates aortic dilation in mice, suggesting that miR-29 may represent a novel molecular target to augment matrix synthesis and maintain vascular wall structural integrity.


Asunto(s)
Aorta/metabolismo , Aneurisma de la Aorta/genética , MicroARNs/análisis , MicroARNs/metabolismo , Envejecimiento/genética , Angiotensina II , Animales , Aorta/patología , Aneurisma de la Aorta/inducido químicamente , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta/patología , Aneurisma de la Aorta/prevención & control , Biopsia , Dilatación Patológica , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Oligorribonucleótidos Antisentido/administración & dosificación
14.
J Am Acad Dermatol ; 68(4): 638-646, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23266341

RESUMEN

BACKGROUND: Episodes of microvascular proliferation associated with volume expansion have been observed in arteriovenous malformations (AVMs) of skin and soft tissue. OBJECTIVE: We sought to investigate the relationship between a microvascular proliferative response and flow velocity in AVMs. METHODS: Resection specimens of 80 AVMs were clinically categorized as either high- or low-flow lesions, and histopathologically screened for the presence of microvessels, inflammation, thrombosis, or a combination of these. Immunohistochemistry was performed with endoglin (CD105), von Willebrand factor, and fibrinogen antibodies. RESULTS: Clinically, 37 AVMs were classified as high-flow lesions and 43 as low-flow lesions. In 81% of high-flow lesions microvascular proliferations were seen versus in 14% of low-flow lesions (P < .005). In high-flow lesions, which were embolized before surgery (30% of all), 88% showed microvascular proliferation, 88% inflammation, and 33% thrombosis. However, similar vasoproliferative responses were also observed in nonembolized AVM (69% high-flow and 14% low-flow lesions). Endoglin was more frequently expressed in high-flow lesions. Extracellular von Willebrand factor staining was found in most lesions, irrespective of flow type or presence of microvascular proliferations. LIMITATIONS: The study was carried out at a single tertiary referral center. CONCLUSIONS: Microvascular proliferative masses in AVMs appear to be strongly associated with high-flow characteristics. This could be explained to some extent by previous therapeutic embolization and/or inflammation in the lesion. However, occurrence of similar microvascular responses in AVM that were not embolized before surgery suggests that the biomechanical effects of high flow in these lesions may also have an angiogenic effect.


Asunto(s)
Malformaciones Arteriovenosas/patología , Malformaciones Arteriovenosas/fisiopatología , Embolización Terapéutica/efectos adversos , Inflamación/complicaciones , Microvasos/patología , Adolescente , Adulto , Anciano , Velocidad del Flujo Sanguíneo , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
15.
Eur Heart J ; 33(9): 1076-84, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21831908

RESUMEN

AIMS: In patients with obstructive coronary artery disease (CAD), the growth of collateral arteries, i.e. arteriogenesis, can preserve myocardial tissue perfusion and function. Monocytes modulate this process, supplying locally the necessary growth factors and degrading enzymes. Knowledge on factors involved in human arteriogenesis is scarce. Thus, the aim of the present study is to identify targets in monocytes that are critical for arteriogenesis in patients with CAD. METHODS AND RESULTS: A total of 50 patients with a chronic total coronary occlusion were dichotomized according to their collateral flow index. From each patient, RNA was isolated from unstimulated peripheral blood monocytes, monocytes stimulated by lipopolysaccharide (LPS) or interleukin (IL)-4, and from macrophages. Increased mRNA expression of galectin-2 was found in three out of four monocytic cell types of patients with a low capacity of the collateral circulation (P= 0.03 for unstimulated monocytes; P= 0.02 for LPS-stimulated monocytes; P= 0.20 for IL-4-stimulated monocytes; P= 0.02 for macrophages). Additionally, galectin-2 mRNA expression was significantly associated with the rs7291467 polymorphism in LGALS2 encoding galectin-2 in all four monocytic cell types. Patient with the rs7291467 CC genotype displayed highest galectin-2 expression, and also tended to have a lower arteriogenic response. To evaluate the effect of galectin-2 on arteriogenesis in vivo, we used a murine hindlimb model. Treatment with galectin-2 markedly impaired the perfusion restoration at Day 7. CONCLUSION: Collectively, these results identify galectin-2 as a novel inhibitor of arteriogenesis. Modulation of galectin-2 may constitute a new therapeutic strategy for the stimulation of arteriogenesis in patients with CAD.


Asunto(s)
Circulación Colateral/genética , Oclusión Coronaria/genética , Galectina 2/metabolismo , Polimorfismo Genético/genética , Anciano , Animales , Fármacos Cardiovasculares/farmacología , Circulación Colateral/efectos de los fármacos , Oclusión Coronaria/metabolismo , Oclusión Coronaria/fisiopatología , Femenino , Galectina 2/genética , Galectina 2/farmacología , Miembro Posterior , Humanos , Interleucina-4/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Monocitos/efectos de los fármacos , ARN Mensajero/metabolismo
16.
J Mol Cell Cardiol ; 52(4): 897-904, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21959171

RESUMEN

Obstruction of coronary arteries leads to an arteriogenic response. Pre-existent collateral networks enlarge, forming large conductance arteries with the capability to compensate for the loss of perfusion due to the occlusion. Interestingly, significant differences exist between patients regarding the capacity to develop such a collateral circulation. This heterogeneity in arteriogenic response is also found between and even within animal species and it strongly suggests that next to environmental factors, innate genetic factors play a key role. The present review focuses on this heterogeneity of genetic as well as non-genetic determinants of the coronary collateral circulation. This article is part of a Special Issue entitled "Coronary Blood Flow".


Asunto(s)
Circulación Colateral/fisiología , Circulación Coronaria/fisiología , Animales , Vasos Coronarios/fisiopatología , Humanos , Modelos Teóricos
17.
J Mol Cell Cardiol ; 53(2): 299-309, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22659288

RESUMEN

High laminar shear stress (HLSS) is vasculoprotective partly through induction of Kruppel-like factor 2 (KLF2). Connexin37 (Cx37) is highly expressed in endothelial cells (ECs) of healthy arteries, but not in ECs overlying atherosclerotic lesions. Moreover, Cx37 deletion in apolipoprotein E-deficient (ApoE(-/-)) mice increases susceptibility to atherosclerosis. We hypothesized that shear stress, through KLF2 modulation, may affect Cx37 expression in ECs. Cx37 expression and gap-junctional intercellular (GJIC) dye transfer are prominent in the straight portion of carotid arteries of ApoE(-/-) mice, but are reduced at the carotid bifurcation, a region subjected to oscillatory flow. Shear stress-modifying vascular casts were placed around the common carotid artery of ApoE(-/-) mice. Whereas Cx37 expression was conserved in HLSS regions, it was downregulated to ~50% in low laminar or oscillatory flow regions. To study the mechanisms involved, HUVECs or bEnd.3 cells were exposed to flow in vitro. Cx37 and KLF2 expression were increased after 24h of HLSS. Interestingly, shear-dependent Cx37 expression was significantly reduced after silencing of KLF2. Moreover after exposure to simvastatin, a well-known KLF2 inducer, KLF2 binds to the Cx37 promoter region as shown by ChIP. Finally, GJIC dye transfer was highly reduced after KLF2 silencing and was increased after exposure to simvastatin. HLSS upregulates the expression of Cx37 in ECs by inducing its transcription factor KLF2, which increases intercellular communication. Therefore, this effect of shear stress on Cx37 expression may contribute to the synchronization of ECs and participate in the protective effect of HLSS.


Asunto(s)
Conexinas/metabolismo , Células Endoteliales/metabolismo , Estrés Mecánico , Animales , Apolipoproteínas E , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Conexinas/genética , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Interferencia de ARN
18.
J Proteome Res ; 11(5): 2925-36, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22468712

RESUMEN

Vascular endothelial cells contain unique storage organelles, designated Weibel-Palade bodies (WPBs), that deliver inflammatory and hemostatic mediators to the vascular lumen in response to agonists like thrombin and vasopressin. The main component of WPBs is von Willebrand factor (VWF), a multimeric glycoprotein crucial for platelet plug formation. In addition to VWF, several other components are known to be stored in WPBs, like osteoprotegerin, monocyte chemoattractant protein-1 and angiopoetin-2 (Ang-2). Here, we used an unbiased proteomics approach to identify additional residents of WPBs. Mass spectrometry analysis of purified WPBs revealed the presence of several known components such as VWF, Ang-2, and P-selectin. Thirty-five novel candidate WPB residents were identified that included insulin-like growth factor binding protein-7 (IGFBP7), which has been proposed to regulate angiogenesis. Immunocytochemistry revealed that IGFBP7 is a bona fide WPB component. Cotransfection studies showed that IGFBP7 trafficked to pseudo-WPB in HEK293 cells. Using a series of deletion variants of VWF, we showed that targeting of IGFBP7 to pseudo-WPBs was dependent on the carboxy-terminal D4-C1-C2-C3-CK domains of VWF. IGFBP7 remained attached to ultralarge VWF strings released upon exocytosis of WPBs under flow. The presence of IGFBP7 in WPBs highlights the role of this subcellular compartment in regulation of angiogenesis.


Asunto(s)
Células Endoteliales/química , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/química , Proteómica/métodos , Cuerpos de Weibel-Palade/química , Células Endoteliales/fisiología , Exocitosis , Vectores Genéticos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunohistoquímica , Espectrometría de Masas , Neovascularización Fisiológica , Selectina-P/química , Estructura Terciaria de Proteína , Transporte de Proteínas , Transfección , Cuerpos de Weibel-Palade/fisiología , Factor de von Willebrand/química
19.
Blood ; 115(12): 2533-42, 2010 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-20032497

RESUMEN

The shear stress-induced transcription factor Krüppel-like factor 2 (KLF2) confers antiinflammatory properties to endothelial cells through the inhibition of activator protein 1, presumably by interfering with mitogen-activated protein kinase (MAPK) cascades. To gain insight into the regulation of these cascades by KLF2, we used antibody arrays in combination with time-course mRNA microarray analysis. No gross changes in MAPKs were detected; rather, phosphorylation of actin cytoskeleton-associated proteins, including focal adhesion kinase, was markedly repressed by KLF2. Furthermore, we demonstrate that KLF2-mediated inhibition of Jun NH(2)-terminal kinase (JNK) and its downstream targets ATF2/c-Jun is dependent on the cytoskeleton. Specifically, KLF2 directs the formation of typical short basal actin filaments, termed shear fibers by us, which are distinct from thrombin- or tumor necrosis factor-alpha-induced stress fibers. KLF2 is shown to be essential for shear stress-induced cell alignment, concomitant shear fiber assembly, and inhibition of JNK signaling. These findings link the specific effects of shear-induced KLF2 on endothelial morphology to the suppression of JNK MAPK signaling in vascular homeostasis via novel actin shear fibers.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Células Endoteliales/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Factor de Transcripción Activador 2/metabolismo , Animales , Aorta/citología , Células Cultivadas , Células Endoteliales/citología , Arteria Femoral/citología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Fosforilación/fisiología , Ratas , Flujo Sanguíneo Regional/fisiología , Vena Safena/citología , Estrés Mecánico , Transducción Genética , Venas Umbilicales/citología , Quinasas Asociadas a rho/metabolismo
20.
Exp Cell Res ; 317(2): 131-42, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20955695

RESUMEN

AIMS: Activin A and transforming growth factor-ß1 (TGF-ß1) belong to the same family of growth and differentiation factors that modulate vascular lesion formation in distinct ways, which we wish to understand mechanistically. METHODS AND RESULTS: We investigated the expression of cell-surface receptors and activation of Smads in human vascular smooth muscle cells (SMCs) and demonstrated that activin receptor-like kinase-1 (ALK-1), ALK-4, ALK-5 and endoglin are expressed in human SMCs. As expected, TGF-ß1 activates Smad1 and Smad2 in these cells. Interestingly, activin A also induces phosphorylation of both Smads, which has not been reported for Smad1 before. Transcriptome analyses of activin A and TGF-ß1 treated SMCs with subsequent Gene-Set Enrichment Analyses revealed that many downstream gene networks are induced by both factors. However, the effect of activin A on expression kinetics of individual genes is less pronounced than for TGF-ß1, which is explained by a more rapid dephosphorylation of Smads and p38-MAPK in response to activin A. Substantial differences in expression of fibronectin, alpha-V integrin and total extracellular collagen synthesis were observed. CONCLUSIONS: Genome-wide mRNA expression analyses clarify the distinct modulation of vascular lesion formation by activin A and TGF-ß1, most significantly because activin A is non-fibrotic.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Activinas/farmacología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Fenotipo , Factor de Crecimiento Transformador beta/farmacología , Receptores de Activinas Tipo I/metabolismo , Activinas/genética , Activinas/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Humanos , Miocitos del Músculo Liso/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Vena Safena/citología , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA