Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Angiogenesis ; 27(1): 23-35, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37326760

RESUMEN

Patients with chronic kidney disease (CKD) have an increased risk for cardiovascular morbidity and mortality. Capillary rarefaction may be both one of the causes as well as a consequence of CKD and cardiovascular disease. We reviewed the published literature on human biopsy studies and conclude that renal capillary rarefaction occurs independently of the cause of renal function decline. Moreover, glomerular hypertrophy may be an early sign of generalized endothelial dysfunction, while peritubular capillary loss occurs in advanced renal disease. Recent studies with non-invasive measurements show that capillary rarefaction is detected systemically (e.g., in the skin) in individuals with albuminuria, as sign of early CKD and/or generalized endothelial dysfunction. Decreased capillary density is found in omental fat, muscle and heart biopsies of patients with advanced CKD as well as in skin, fat, muscle, brain and heart biopsies of individuals with cardiovascular risk factors. No biopsy studies have yet been performed on capillary rarefaction in individuals with early CKD. At present it is unknown whether individuals with CKD and cardiovascular disease merely share the same risk factors for capillary rarefaction, or whether there is a causal relationship between rarefaction in renal and systemic capillaries. Further studies on renal and systemic capillary rarefaction, including their temporal relationship and underlying mechanisms are needed. This review stresses the importance of preserving and maintaining capillary integrity and homeostasis in the prevention and management of renal and cardiovascular disease.


Asunto(s)
Enfermedades Cardiovasculares , Rarefacción Microvascular , Insuficiencia Renal Crónica , Enfermedades Vasculares , Humanos , Capilares/patología , Enfermedades Cardiovasculares/patología , Rarefacción Microvascular/patología , Riñón/patología , Insuficiencia Renal Crónica/patología , Enfermedades Vasculares/patología
2.
Angiogenesis ; 27(3): 461-474, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38780883

RESUMEN

The presence of atherosclerotic plaque vessels is a critical factor in plaque destabilization. This may be attributable to the leaky phenotype of these microvessels, although direct proof for this notion is lacking. In this study, we investigated molecular and cellular patterns of stable and hemorrhaged human plaque to identify novel drivers of intraplaque vessel dysfunction. From transcriptome data of a human atherosclerotic lesion cohort, we reconstructed a co-expression network, identifying a gene module strongly and selectively correlated with both plaque microvascular density and inflammation. Spectrin Beta Non-Erythrocytic 1 (sptbn1) was identified as one of the central hubs of this module (along with zeb1 and dock1) and was selected for further study based on its predominant endothelial expression. Silencing of sptbn1 enhanced leukocyte transmigration and vascular permeability in vitro, characterized by an increased number of focal adhesions and reduced junctional VE-cadherin. In vivo, sptbn1 knockdown in zebrafish impaired the development of the caudal vein plexus. Mechanistically, increased substrate stiffness was associated with sptbn1 downregulation in endothelial cells in vitro and in human vessels. Plaque SPTBN1 mRNA and protein expression were found to correlate with an enhanced presence of intraplaque hemorrhage and future cardiovascular disease (CVD) events during follow-up. In conclusion, we identify SPTBN1 as a central hub gene in a gene program correlating with plaque vascularisation. SPTBN1 was regulated by substrate stiffness in vitro while silencing blocked vascular development in vivo, and compromised barrier function in vitro. Together, SPTBN1 is identified as a new potential regulator of the leaky phenotype of atherosclerotic plaque microvessels.


Asunto(s)
Microvasos , Fenotipo , Placa Aterosclerótica , Espectrina , Pez Cebra , Humanos , Placa Aterosclerótica/patología , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Pez Cebra/genética , Animales , Microvasos/patología , Microvasos/metabolismo , Espectrina/genética , Espectrina/metabolismo , Transcriptoma/genética , Permeabilidad Capilar/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo
3.
Neth Heart J ; 31(12): 461-470, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37910335

RESUMEN

BACKGROUND: Approximately one-third of patients with symptomatic severe aortic valve stenosis who are scheduled for transcatheter aortic valve implantation (TAVI) have some degree of cognitive impairment. TAVI may have negative cognitive effects due to periprocedural micro-emboli inducing cerebral infarction. On the contrary, TAVI may also have positive cognitive effects due to increases in cardiac output and cerebral blood flow (CBF). However, studies that systematically assess these effects are scarce. Therefore, the main aim of this study is to assess cerebral and cognitive outcomes in patients with severe aortic valve stenosis undergoing TAVI. STUDY DESIGN: In the prospective CAPITA (CArdiac OutPut, Cerebral Blood Flow and Cognition In Patients With Severe Aortic Valve Stenosis Undergoing Transcatheter Aortic Valve Implantation) study, cerebral and cognitive outcomes are assessed in patients undergoing TAVI. One day before and 3 months after TAVI, patients will undergo echocardiography (cardiac output, valve function), brain magnetic resonance imaging (CBF, structural lesions) and extensive neuropsychological assessment. To assess longer-term effects of TAVI, patients will again undergo echocardiography and neuropsychological assessment 1 year after the procedure. The co-primary outcome measures are change in CBF (in ml/100 g per min) and change in global cognitive functioning (Z-score) between baseline and 3­month follow-up. Secondary objectives include change in cardiac output, white matter hyperintensities and other structural brain lesions. (ClinicalTrials.gov identifier NCT05481008) CONCLUSION : The CAPITA study is the first study designed to systematically assess positive and negative cerebral and cognitive outcomes after TAVI. We hypothesise that TAVI improves cardiac output, CBF and cognitive functioning.

5.
Eur Heart J ; 40(4): 372-382, 2019 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-30452556

RESUMEN

Aims: The E3-ligase CBL-B (Casitas B-cell lymphoma-B) is an important negative regulator of T cell activation that is also expressed in macrophages. T cells and macrophages mediate atherosclerosis, but their regulation in this disease remains largely unknown; thus, we studied the function of CBL-B in atherogenesis. Methods and results: The expression of CBL-B in human atherosclerotic plaques was lower in advanced lesions compared with initial lesions and correlated inversely with necrotic core area. Twenty weeks old Cblb-/-Apoe-/- mice showed a significant increase in plaque area in the aortic arch, where initial plaques were present. In the aortic root, a site containing advanced plaques, lesion area rose by 40%, accompanied by a dramatic change in plaque phenotype. Plaques contained fewer macrophages due to increased apoptosis, larger necrotic cores, and more CD8+ T cells. Cblb-/-Apoe-/- macrophages exhibited enhanced migration and increased cytokine production and lipid uptake. Casitas B-cell lymphoma-B deficiency increased CD8+ T cell numbers, which were protected against apoptosis and regulatory T cell-mediated suppression. IFNγ and granzyme B production was enhanced in Cblb-/-Apoe-/- CD8+ T cells, which provoked macrophage killing. Depletion of CD8+ T cells in Cblb-/-Apoe-/- bone marrow chimeras rescued the phenotype, indicating that CBL-B controls atherosclerosis mainly through its function in CD8+ T cells. Conclusion: Casitas B-cell lymphoma-B expression in human plaques decreases during the progression of atherosclerosis. As an important regulator of immune responses in experimental atherosclerosis, CBL-B hampers macrophage recruitment and activation during initial atherosclerosis and limits CD8+ T cell activation and CD8+ T cell-mediated macrophage death in advanced atherosclerosis, thereby preventing the progression towards high-risk plaques.


Asunto(s)
Aterosclerosis/etiología , Linfocitos T CD8-positivos/inmunología , Linfoma de Células B/complicaciones , Macrófagos/patología , Proteína Oncogénica v-cbl/metabolismo , Placa Aterosclerótica/etiología , Animales , Apoptosis , Aterosclerosis/metabolismo , Aterosclerosis/patología , Modelos Animales de Enfermedad , Humanos , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología
6.
Circ Res ; 121(6): e53-e79, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28729353

RESUMEN

Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.


Asunto(s)
American Heart Association , Aterosclerosis/fisiopatología , Guías de Práctica Clínica como Asunto , Proyectos de Investigación/normas , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Investigación Biomédica/normas , Estados Unidos
7.
J Cardiovasc Magn Reson ; 21(1): 15, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30832656

RESUMEN

BACKGROUND: The presence of intraplaque haemorrhage (IPH) has been related to plaque rupture, is associated with plaque progression, and predicts cerebrovascular events. However, the mechanisms leading to IPH are not fully understood. The dominant view is that IPH is caused by leakage of erythrocytes from immature microvessels. The aim of the present study was to investigate whether there is an association between atherosclerotic plaque microvasculature and presence of IPH in a relatively large prospective cohort study of patients with symptomatic carotid plaque. METHODS: One hundred and thirty-two symptomatic patients with ≥2 mm carotid plaque underwent cardiovascular magnetic resonance (CMR) of the symptomatic carotid plaque for detection of IPH and dynamic contrast-enhanced (DCE)-CMR for assessment of plaque microvasculature. Ktrans, an indicator of microvascular flow, density and leakiness, was estimated using pharmacokinetic modelling in the vessel wall and adventitia. Statistical analysis was performed using an independent samples T-test and binary logistic regression, correcting for clinical risk factors. RESULTS: A decreased vessel wall Ktrans was found for IPH positive patients (0.051 ± 0.011 min- 1 versus 0.058 ± 0.017 min- 1, p = 0.001). No significant difference in adventitial Ktrans was found in patients with and without IPH (0.057 ± 0.012 min- 1 and 0.057 ± 0.018 min- 1, respectively). Histological analysis in a subgroup of patients that underwent carotid endarterectomy demonstrated no significant difference in relative microvessel density between plaques without IPH (n = 8) and plaques with IPH (n = 15) (0.000333 ± 0.0000707 vs. and 0.000289 ± 0.0000439, p = 0.585). CONCLUSIONS: A reduced vessel wall Ktrans is found in the presence of IPH. Thus, we did not find a positive association between plaque microvasculature and IPH several weeks after a cerebrovascular event. Not only leaky plaque microvessels, but additional factors may contribute to IPH development. TRIAL REGISTRATION: NCT01208025 . Registration date September 23, 2010. Retrospectively registered (first inclusion September 21, 2010). NCT01709045 , date of registration October 17, 2012. Retrospectively registered (first inclusion August 23, 2011).


Asunto(s)
Arterias Carótidas/diagnóstico por imagen , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Hemorragia/diagnóstico por imagen , Imagen por Resonancia Magnética , Microvasos/diagnóstico por imagen , Placa Aterosclerótica , Anciano , Arterias Carótidas/patología , Arterias Carótidas/cirugía , Enfermedades de las Arterias Carótidas/complicaciones , Enfermedades de las Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/cirugía , Medios de Contraste/administración & dosificación , Endarterectomía Carotidea , Hemorragia/patología , Humanos , Ataque Isquémico Transitorio/etiología , Masculino , Microvasos/patología , Persona de Mediana Edad , Compuestos Organometálicos/administración & dosificación , Valor Predictivo de las Pruebas , Estudios Prospectivos , Factores de Riesgo , Accidente Cerebrovascular/etiología
8.
Arterioscler Thromb Vasc Biol ; 37(9): e131-e157, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28729366

RESUMEN

Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.


Asunto(s)
American Heart Association , Aterosclerosis , Investigación Biomédica/normas , Recolección de Datos/normas , Proyectos de Investigación/normas , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Primates , Conejos , Especificidad de la Especie , Porcinos , Estados Unidos
9.
J Magn Reson Imaging ; 46(4): 1053-1059, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28152245

RESUMEN

PURPOSE: To assess parameter agreement of volume transfer coefficient (Ktrans ) between two vascular regions and to study the correlation with microvessel density on histology. The dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) parameter Ktrans is frequently used to study atherosclerotic plaque microvasculature. Ktrans has been reported using different descriptive statistics (mean, median, 75th percentile) either for the whole vessel wall or the adventitia in previous studies. MATERIALS AND METHODS: DCE-MRI parameter agreement was analyzed in 110 symptomatic patients with ≥2 mm carotid plaque that underwent a 3T carotid DCE-MRI examination. Ktrans was estimated in the entire vessel wall and adventitia. Twenty-three patients underwent carotid endarterectomy and were used for comparison with histological quantification of microvessel density of the plaque using CD31 immunohistochemistry. DCE-MRI parameters in the vessel wall regions were compared using Pearson's correlation coefficient, Bland-Altman analysis, and a two-sided paired samples t-test. Correlation of the DCE-MRI parameters with histology was studied using the Pearson's correlation coefficient. RESULTS: Median adventitial Ktrans was 5% higher (P = 0.003) than entire vessel wall Ktrans , with no differences for other descriptive statistics. Vessel wall and adventitial Ktrans showed similar moderately strong correlations with plaque microvessel density on histology (Pearson's ρ: 0.59-0.65 [P < 0.003] and 0.52-0.64 [P < 0.011], respectively). CONCLUSION: The similar moderately strong correlations for vessel wall and adventitial Ktrans with microvessel density on histology suggested that both regions reflected plaque microvessel density. Care should to be taken when comparing absolute values between studies. Future studies incorporating thresholds for risk stratification need to agree upon standardization of DCE-MRI parameters. LEVEL OF EVIDENCE: 2 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2017;46:1053-1059.


Asunto(s)
Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Medios de Contraste/farmacocinética , Aumento de la Imagen/métodos , Imagen por Resonancia Magnética/métodos , Microvasos/diagnóstico por imagen , Placa Aterosclerótica/diagnóstico por imagen , Anciano , Arterias Carótidas/diagnóstico por imagen , Estudios Transversales , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Masculino , Estudios Prospectivos
10.
Circ Res ; 116(5): 895-908, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25722443

RESUMEN

Vascular stiffness is a mechanical property of the vessel wall that affects blood pressure, permeability, and inflammation. As a result, vascular stiffness is a key driver of (chronic) human disorders, including pulmonary arterial hypertension, kidney disease, and atherosclerosis. Responses of the endothelium to stiffening involve integration of mechanical cues from various sources, including the extracellular matrix, smooth muscle cells, and the forces that derive from shear stress of blood. This response in turn affects endothelial cell contractility, which is an important property that regulates endothelial stiffness, permeability, and leukocyte-vessel wall interactions. Moreover, endothelial stiffening reduces nitric oxide production, which promotes smooth muscle cell contraction and vasoconstriction. In fact, vessel wall stiffening, and microcirculatory endothelial dysfunction, precedes hypertension and thus underlies the development of vascular disease. Here, we review the cross talk among vessel wall stiffening, endothelial contractility, and vascular disease, which is controlled by Rho-driven actomyosin contractility and cellular mechanotransduction. In addition to discussing the various inputs and relevant molecular events in the endothelium, we address which actomyosin-regulated changes at cell adhesion complexes are genetically associated with human cardiovascular disease. Finally, we discuss recent findings that broaden therapeutic options for targeting this important mechanical signaling pathway in vascular pathogenesis.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Endotelio Vascular/fisiopatología , Mecanotransducción Celular/fisiología , Rigidez Vascular/fisiología , Quinasas Asociadas a rho/fisiología , Actomiosina/fisiología , Envejecimiento/fisiología , Animales , Calcinosis/patología , Calcinosis/fisiopatología , Enfermedades Cardiovasculares/enzimología , Adhesión Celular/fisiología , Permeabilidad de la Membrana Celular , Citoesqueleto/ultraestructura , Endotelio Vascular/ultraestructura , Hemorreología , Humanos , Inflamación , Integrinas/fisiología , Leucocitos/fisiología , Mecanotransducción Celular/efectos de los fármacos , Ratones , Ratones Noqueados , Microcirculación , Modelos Cardiovasculares , Fosfatasa de Miosina de Cadena Ligera/antagonistas & inhibidores , Fosfatasa de Miosina de Cadena Ligera/fisiología , FN-kappa B/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Análisis de la Onda del Pulso , Ratas , Migración Transendotelial y Transepitelial , Rigidez Vascular/efectos de los fármacos , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/fisiología , Quinasas Asociadas a rho/antagonistas & inhibidores
11.
Eur Heart J ; 37(39): 2993-2997, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27125949

RESUMEN

AIMS: Normalization of hypercholesterolaemia, inflammation, hyperglycaemia, and obesity are main desired targets to prevent cardiovascular clinical events. Here we present a novel regulator of cholesterol metabolism, which simultaneously impacts on glucose intolerance and inflammation. METHODS AND RESULTS: Mice deficient for oxygen sensor HIF-prolyl hydroxylase 1 (PHD1) were backcrossed onto an atherogenic low-density lipoprotein receptor (LDLR) knockout background and atherosclerosis was studied upon 8 weeks of western-type diet. PHD1-/-LDLR-/- mice presented a sharp reduction in VLDL and LDL plasma cholesterol levels. In line, atherosclerotic plaque development, as measured by plaque area, necrotic core expansion and plaque stage was hampered in PHD1-/-LDLR-/- mice. Mechanistically, cholesterol-lowering in PHD1 deficient mice was a result of enhanced cholesterol excretion from blood to intestines and ultimately faeces. Additionally, flow cytometry of whole blood of these mice revealed significantly reduced counts of leucocytes and particularly of Ly6Chigh pro-inflammatory monocytes. In addition, when studying PHD1-/- in diet-induced obesity (14 weeks high-fat diet) mice were less glucose intolerant when compared with WT littermate controls. CONCLUSION: Overall, PHD1 knockout mice display a metabolic phenotype that generally is deemed protective for cardiovascular disease. Future studies should focus on the efficacy, safety, and gender-specific effects of PHD1 inhibition in humans, and unravel the molecular actors responsible for PHD1-driven, likely intestinal, and regulation of cholesterol metabolism.


Asunto(s)
Aterosclerosis , Hipercolesterolemia , Hiperglucemia , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno , Prolil Hidroxilasas , Receptores de LDL
12.
Curr Opin Lipidol ; 26(5): 405-11, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26241102

RESUMEN

PURPOSE OF REVIEW: Neovascularization is a prominent feature in advanced human atherosclerotic plaques. This review surveys recent evidence for and remaining uncertainties regarding a role of neovascularization in atherosclerotic plaque progression. Specific emphasis is given to hypoxia, angiogenesis inhibition, and perivascular adipose tissue (PVAT). RECENT FINDINGS: Immunohistochemical and imaging studies showed a strong association between hypoxia, inflammation and neovascularization, and the progression of the atherosclerotic plaque both in humans and mice. Whereas in humans, a profound invasion of microvessels from the adventitia into the plaque occurs, neovascularization in mice is found mainly (peri)adventitially. Influencing neovascularization in mice affected plaque progression, possibly by improving vessel perfusion, but supportive clinical data are not available. Whereas plaque neovascularization contributes to monocyte/macrophage accumulation in the plaque, lymphangiogenesis may facilitate egress of cells and waste products. A specific role for PVAT and its secreted factors is anticipated and wait further clinical evaluation. SUMMARY: Hypoxia, inflammation, and plaque neovascularization are associated with plaque progression as underpinned by recent imaging data in humans. Recent studies provide new insights into modulation of adventitia-associated angiogenesis, PVAT, and plaque development in mice, but there is still a need for detailed information on modulating human plaque vascularization in patients.


Asunto(s)
Adventicia/irrigación sanguínea , Aterosclerosis/fisiopatología , Microvasos/patología , Neovascularización Patológica/patología , Placa Aterosclerótica/fisiopatología , Adiponectina/fisiología , Tejido Adiposo/patología , Adventicia/patología , Animales , Aterosclerosis/sangre , Aterosclerosis/inmunología , Hipoxia de la Célula , Humanos , Neovascularización Patológica/sangre , Neovascularización Patológica/inmunología , Oxígeno/sangre , Placa Aterosclerótica/patología
13.
Stroke ; 46(12): 3411-5, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26514191

RESUMEN

BACKGROUND AND PURPOSE: Intraplaque hemorrhage (IPH), visualized by magnetic resonance imaging, has shown to be associated with the risk of stroke in patients with carotid artery stenosis. The mechanisms of IPH development are poorly understood. In this study, we investigated the association between clinical patient characteristics and carotid IPH on high-resolution magnetic resonance imaging. METHODS: Patients participate in the Plaque at Risk (PARISK) study. This prospective, multicenter cohort study included patients with recent amaurosis fugax, hemispheric transient ischemic attack, or nondisabling stroke in the internal carotid artery territory and an ipsilateral carotid stenosis of <70%, who were not scheduled for carotid revascularization procedure. One hundred patients, recruited between 2010 and 2012, underwent a 3-T high-resolution carotid magnetic resonance imaging. We documented clinical patient characteristics and performed multivariable logistic regression analysis to investigate their association with IPH. RESULTS: IPH was observed in 45 patients (45%) in 1 or both carotid arteries. Male sex and the use of antiplatelet agents before the index event were associated with IPH in univariable analysis. In a multivariable analysis, only previous use of antiplatelet agents was significantly associated with IPH (odds ratio, 2.71; 95% confidence interval, 1.12-6.61). Risk factors of atherosclerotic arterial disease, including a history of symptomatic arterial diseases, were not associated with IPH. CONCLUSIONS: In this cohort of 100 patients with recently symptomatic carotid stenosis, the previous use of antiplatelet agents is associated with carotid IPH on magnetic resonance imaging. Antiplatelet therapy may increase the risk of IPH, but our findings need to be confirmed in larger patient cohorts. The implications for risk stratification remain to be determined.


Asunto(s)
Arterias Carótidas/patología , Estenosis Carotídea/diagnóstico , Hemorragia/inducido químicamente , Hemorragia/diagnóstico , Placa Aterosclerótica/diagnóstico , Inhibidores de Agregación Plaquetaria/efectos adversos , Anciano , Arterias Carótidas/metabolismo , Estenosis Carotídea/tratamiento farmacológico , Estenosis Carotídea/metabolismo , Estudios de Cohortes , Estudios Transversales , Femenino , Hemorragia/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/metabolismo , Estudios Prospectivos , Factores de Riesgo
14.
Arterioscler Thromb Vasc Biol ; 34(9): 2059-67, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25012130

RESUMEN

OBJECTIVE: Vascular endothelial-cadherin- and integrin-based cell adhesions are crucial for endothelial barrier function. Formation and disassembly of these adhesions controls endothelial remodeling during vascular repair, angiogenesis, and inflammation. In vitro studies indicate that vascular cytokines control adhesion through regulation of the actin cytoskeleton, but it remains unknown whether such regulation occurs in human vessels. We aimed to investigate regulation of the actin cytoskeleton and cell adhesions within the endothelium of human arteries and veins. APPROACH AND RESULTS: We used an ex vivo protocol for immunofluorescence in human vessels, allowing detailed en face microscopy of endothelial monolayers. We compared arteries and veins of the umbilical cord and mesenteric, epigastric, and breast tissues and find that the presence of central F-actin fibers distinguishes the endothelial phenotype of adult arteries from veins. F-actin in endothelium of adult veins as well as in umbilical vasculature predominantly localizes cortically at the cell boundaries. By contrast, prominent endothelial F-actin fibers in adult arteries anchor mostly to focal adhesions containing integrin-binding proteins paxillin and focal adhesion kinase and follow the orientation of the extracellular matrix protein fibronectin. Other arterial F-actin fibers end in vascular endothelial-cadherin-based endothelial focal adherens junctions. In vitro adhesion experiments on compliant substrates demonstrate that formation of focal adhesions is strongly induced by extracellular matrix rigidity, irrespective of arterial or venous origin of endothelial cells. CONCLUSIONS: Our data show that F-actin-anchored focal adhesions distinguish endothelial phenotypes of human arteries from veins. We conclude that the biomechanical properties of the vascular extracellular matrix determine this endothelial characteristic.


Asunto(s)
Actinas/análisis , Arterias/citología , Endotelio Vascular/citología , Adhesiones Focales , Venas/citología , Citoesqueleto de Actina/ultraestructura , Adulto , Antígenos CD/análisis , Mama/irrigación sanguínea , Cadherinas/análisis , Células Cultivadas , Células Endoteliales , Arterias Epigástricas/citología , Matriz Extracelular/fisiología , Proteínas de la Matriz Extracelular/fisiología , Femenino , Humanos , Recién Nacido , Arterias Mesentéricas/citología , Venas Mesentéricas/citología , Microscopía Confocal , Microscopía Fluorescente , Fenotipo , Arterias Umbilicales/citología , Venas Umbilicales/citología
15.
Arterioscler Thromb Vasc Biol ; 34(12): 2545-53, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25256233

RESUMEN

OBJECTIVE: Advanced murine and human plaques are hypoxic, but it remains unclear whether plaque hypoxia is causally related to atherogenesis. Here, we test the hypothesis that reversal of hypoxia in atherosclerotic plaques by breathing hyperoxic carbogen gas will prevent atherosclerosis. APPROACH AND RESULTS: Low-density lipoprotein receptor-deficient mice (LDLR(-/-)) were fed a Western-type diet, exposed to carbogen (95% O2, 5% CO2) or air, and the effect on plaque hypoxia, size, and phenotype was studied. First, the hypoxic marker pimonidazole was detected in murine LDLR(-/-) plaque macrophages from plaque initiation onwards. Second, the efficacy of breathing carbogen (90 minutes, single exposure) was studied. Compared with air, carbogen increased arterial blood pO2 5-fold in LDLR(-/-) mice and reduced plaque hypoxia in advanced plaques of the aortic root (-32%) and arch (-84%). Finally, the effect of repeated carbogen exposure on progression of atherosclerosis was studied in LDLR(-/-) mice fed a Western-type diet for an initial 4 weeks, followed by 4 weeks of diet and carbogen or air (both 90 min/d). Carbogen reduced plaque hypoxia (-40%), necrotic core size (-37%), and TUNEL(+) (terminal uridine nick-end labeling positive) apoptotic cell content (-50%) and increased efferocytosis of apoptotic cells by cluster of differentiation 107b(+) (CD107b, MAC3) macrophages (+36%) in advanced plaques of the aortic root. Plaque size, plasma cholesterol, hematopoiesis, and systemic inflammation were unchanged. In vitro, hypoxia hampered efferocytosis by bone marrow-derived macrophages, which was dependent on the receptor Mer tyrosine kinase. CONCLUSIONS: Carbogen restored murine plaque oxygenation and prevented necrotic core expansion by enhancing efferocytosis, likely via Mer tyrosine kinase. Thus, plaque hypoxia is causally related to necrotic core expansion.


Asunto(s)
Hipoxia/patología , Placa Aterosclerótica/patología , Placa Aterosclerótica/prevención & control , Animales , Apoptosis , Antígenos CD36/deficiencia , Antígenos CD36/genética , Dióxido de Carbono/administración & dosificación , Humanos , Hipoxia/fisiopatología , Hipoxia/terapia , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis , Oxígeno/administración & dosificación , Oxígeno/sangre , Fagocitosis , Placa Aterosclerótica/fisiopatología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Tirosina Quinasas Receptoras/deficiencia , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Tirosina Quinasa c-Mer
16.
Eur Heart J ; 35(17): 1137-46, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24126878

RESUMEN

AIMS: Rupture-prone atherosclerotic plaques are characterized by inflammation and a large necrotic core. Inflammation is linked to high metabolic activity. Advanced glycation endproducts (AGEs) and their major precursor methylglyoxal are formed during high metabolic activity and can have detrimental effects on cellular function and may induce cell death. Therefore, we investigated whether plaque AGEs are increased in human carotid rupture-prone plaques and are associated with plaque inflammation and necrotic core formation. METHODS AND RESULTS: The protein-bound major methylglyoxal-derived AGE 5-hydro-5-methylimidazolone (MG-H1) and N(ε)-(carboxymethyl)lysine (CML) were measured in human carotid endarterectomy specimens (n = 75) with tandem mass spectrometry. MG-H1 and CML levels were associated with rupture-prone plaques, increased protein levels of the inflammatory mediators IL-8 and MCP-1 and with higher MMP-9 activity. Immunohistochemistry showed that AGEs accumulated predominantly in macrophages surrounding the necrotic core and co-localized with cleaved caspase-3. Intra-plaque comparison revealed that glyoxalase-1 (GLO-1), the major methylglyoxal-detoxifying enzyme, mRNA was decreased (-13%, P < 0.05) in ruptured compared with stable plaque segments. In line, in U937 monoctyes, we found reduced (GLO-1) activity (-38%, P < 0.05) and increased MGO (346%, P < 0.05) production after stimulation with the inflammatory mediator TNF. Direct incubation with methylglyoxal increased apoptosis up to two-fold. CONCLUSION: This is the first study showing that AGEs are associated with human rupture-prone plaques. Furthermore, this study suggests a cascade linking inflammation, reduced GLO-1, methylglyoxal- and AGE-accumulation, and subsequent apoptosis. Thereby, AGEs may act as mediators of the progression of stable to rupture-prone plaques, opening a window towards novel treatments and biomarkers to treat cardiovascular diseases.


Asunto(s)
Aneurisma Roto/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Placa Aterosclerótica/metabolismo , Anciano , Animales , Apoptosis/efectos de los fármacos , Hipoxia de la Célula/fisiología , Humanos , Leucocitos Mononucleares/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Fenotipo , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
17.
Eur Heart J ; 35(43): 3013-20, 3020a-3020d, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25230814

RESUMEN

Blood vessels are exposed to multiple mechanical forces that are exerted on the vessel wall (radial, circumferential and longitudinal forces) or on the endothelial surface (shear stress). The stresses and strains experienced by arteries influence the initiation of atherosclerotic lesions, which develop at regions of arteries that are exposed to complex blood flow. In addition, plaque progression and eventually plaque rupture is influenced by a complex interaction between biological and mechanical factors-mechanical forces regulate the cellular and molecular composition of plaques and, conversely, the composition of plaques determines their ability to withstand mechanical load. A deeper understanding of these interactions is essential for designing new therapeutic strategies to prevent lesion development and promote plaque stabilization. Moreover, integrating clinical imaging techniques with finite element modelling techniques allows for detailed examination of local morphological and biomechanical characteristics of atherosclerotic lesions that may be of help in prediction of future events. In this ESC Position Paper on biomechanical factors in atherosclerosis, we summarize the current 'state of the art' on the interface between mechanical forces and atherosclerotic plaque biology and identify potential clinical applications and key questions for future research.


Asunto(s)
Arterias/fisiología , Aterosclerosis/fisiopatología , Apoptosis/fisiología , Biomarcadores/metabolismo , Fenómenos Biomecánicos/fisiología , Proliferación Celular/fisiología , Senescencia Celular/fisiología , Progresión de la Enfermedad , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Homeostasis/fisiología , Humanos , Mecanorreceptores/fisiología , Placa Aterosclerótica/fisiopatología , Rotura Espontánea/fisiopatología , Transducción de Señal/fisiología , Estrés Mecánico , Remodelación Vascular/fisiología
18.
Stroke ; 45(11): 3423-6, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25256179

RESUMEN

BACKGROUND AND PURPOSE: In patients with mild to moderate symptomatic carotid artery stenosis, intraplaque hemorrhage (IPH) and a thin/ruptured fibrous cap (FC) as evaluated with MRI, and the presence of microembolic signals (MESs) as detected with transcranial Doppler, are associated with an increased risk of a (recurrent) stroke. The objective of the present study is to determine whether the prevalence of MES differs in patients with and without IPH and thin/ruptured FC, and patients with only a thin/ruptured FC without IPH. METHODS: In this multicenter, diagnostic cohort study, patients with recent transient ischemic attack or minor stroke in the carotid territory and an ipsilateral mild to moderate carotid artery plaque were included. IPH and FC status were dichotomously scored. Analysis of transcranial Doppler data was done blinded for the MRI results. Differences between groups were analyzed with Fisher exact test. RESULTS: A total of 113 patients were included. Transcranial Doppler measurements were feasible in 105 patients (average recording time, 219 minutes). A total of 26 MESs were detected in 8 of 105 patients. In 44 of 105 plaques IPH was present. In 92 of 105 plaques FC status was assessable, 36 of these had a thin/ruptured FC. No significant difference in the prevalence of MES between patients with and without IPH (P=0.46) or with thick versus thin/ruptured FC (P=0.48) was found. CONCLUSIONS: In patients with a symptomatic mild to moderate carotid artery stenosis, IPH and FC status are not associated with MES. This suggests that MRI and transcranial Doppler provide different information on plaque vulnerability. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01709045.


Asunto(s)
Estenosis Carotídea/diagnóstico , Hemorragia Cerebral/diagnóstico , Embolia Intracraneal/diagnóstico , Microcirculación , Placa Aterosclerótica/diagnóstico , Anciano , Estenosis Carotídea/epidemiología , Hemorragia Cerebral/epidemiología , Estudios de Cohortes , Femenino , Humanos , Embolia Intracraneal/epidemiología , Masculino , Microcirculación/fisiología , Persona de Mediana Edad , Placa Aterosclerótica/epidemiología , Método Simple Ciego
19.
Arterioscler Thromb Vasc Biol ; 33(2): 249-56, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23241413

RESUMEN

OBJECTIVE: Neovascularization of human atherosclerotic plaques is implicated in plaque progression and destabilization, although its functional implications are yet unresolved. Here, we aimed to elucidate functional and morphological properties of plaque microvessels in mice in vivo. METHODS AND RESULTS: Atherosclerotic carotid arteries from aged (>40 weeks) apolipoprotein E-deficient mice were imaged in vivo using multiphoton laser scanning microscopy. Two distinct groups of vasa vasorum microvessels were observed at sites of atherosclerosis development (median diameters of 18.5 and 5.9 µm, respectively), whereas microvessels within the plaque could only rarely be found. In vivo imaging showed ongoing angiogenic activity and injection of fluorescein isothiocyanate-dextran confirmed active perfusion. Plaque vasa vasorum showed increased microvascular leakage, combined with a loss of endothelial glycocalyx. Mean blood flow velocity in plaque-associated vasa vasorum was reduced by ±50% compared with diameter-matched control capillaries, whereas mean blood flow was reduced 8-fold. Leukocyte adhesion and extravasation were increased 6-fold in vasa vasorum versus control capillaries. CONCLUSIONS: Using a novel in vivo functional imaging strategy, we showed that plaque-associated vasa vasorum were angiogenically active and, albeit poorly, perfused. Moreover, plaque-associated vasa vasorum showed increased permeability, reduced blood flow, and increased leukocyte adhesion and extravasation (ie, characteristics that could contribute to plaque progression and destabilization).


Asunto(s)
Envejecimiento/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Arterias Carótidas/metabolismo , Microvasos/metabolismo , Vasa Vasorum/metabolismo , Factores de Edad , Envejecimiento/genética , Envejecimiento/patología , Animales , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Velocidad del Flujo Sanguíneo , Permeabilidad Capilar , Arterias Carótidas/inmunología , Arterias Carótidas/patología , Arterias Carótidas/fisiopatología , Adhesión Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Leucocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación , Microscopía Confocal , Microscopía de Fluorescencia por Excitación Multifotónica , Microvasos/inmunología , Microvasos/patología , Microvasos/fisiopatología , Neovascularización Patológica , Placa Aterosclerótica , Flujo Sanguíneo Regional , Factores de Tiempo , Vasa Vasorum/inmunología , Vasa Vasorum/patología , Vasa Vasorum/fisiopatología
20.
Eur Heart J ; 34(48): 3717-27, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22613345

RESUMEN

AIMS: The importance of transforming growth factor beta (TGFß) as an immune regulatory cytokine in atherosclerosis has been established. However, the role of TGFß signalling in dendritic cells (DCs) and in DC-mediated T cell proliferation and differentiation in atherosclerosis is unknown. METHODS AND RESULTS: Here, we investigated the effect of disrupted TGFß signalling in DCs on atherosclerosis by using mice carrying a transgene resulting in functional inactivation of TGFß receptor II (TGFßRII) signalling in CD11c(+) cells (Apoe(-/-)CD11cDNR). Apoe(-/-)CD11cDNR mice exhibited an over two-fold increase in the plaque area compared with Apoe(-/-) mice. Plaques of Apoe(-/-)CD11cDNR mice showed an increase in CD45(+) leucocyte content, and specifically in CD3(+), CD4(+) and CD8(+) cells, whereas macrophage content was not affected. In lymphoid organs, Apoe(-/-)CD11cDNR mice had equal amounts of CD11c(+) cells, and CD11c(+)CD8(+) and CD11c(+)CD8(-) subsets, but showed a subtle shift in the CD11c(+)CD8(-) population towards the more inflammatory CD11c(+)CD8(-)CD4(-) DC subset. In addition, the number of plasmacytoid-DCs decreased. Maturation markers such as MHCII, CD86 and CD40 on CD11c(hi) cells did not change, but the CD11cDNR DCs produced more TNFα and IL-12. CD11c(+) cells from CD11cDNR mice strongly induced T-cell proliferation and activation, resulting in increased amounts of effector T cells producing high amounts of Th1 (IFN-γ), Th2 (IL-4, IL-10), Th17 (IL-17), and Treg (IL-10) cytokines. CONCLUSION: Here, we show that loss of TGFßRII signalling in CD11c(+) cells induces subtle changes in DC subsets, which provoke uncontrolled T cell activation and maturation. This results in increased atherosclerosis and an inflammatory plaque phenotype during hypercholesterolaemia.


Asunto(s)
Aterosclerosis/inmunología , Células Dendríticas/inmunología , Inmunidad Celular/inmunología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Linfocitos T/inmunología , Animales , Antígenos CD11/inmunología , Células Cultivadas , Hipercolesterolemia/inmunología , Ratones , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/deficiencia , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA