Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38946422

RESUMEN

Type 2 diabetes (T2D) constitutes a major public health problem, and despite prevention efforts, this pandemic disease is 'one of the deadliest diseases in the world. In 2022, 6.7 million T2D patients died prematurely from vascular complications. Indeed, diabetes increases the risk of myocardial infarction or stroke eightfold. The identification of the molecular actors involved in the occurrence of cardiovascular complications and their prevention are therefore major axes. Our hypothesis is that factors brought into play during physiological aging appear prematurely with diabetes progression. Our study focused on the aging of the extracellular matrix (ECM), a major element in the maintenance of vascular homeostasis. We characterized the morphological and functional aspects of aorta, with a focus on the collagen and elastic fibers of diabetic mice aged from 6 months to non-diabetic mice aged 6 months and 20 months. The comparison with the two non-diabetic models (young and old) highlighted an exacerbated activity of proteases, which could explain a disturbance in the collagen accumulation and an excessive degradation of elastic fibers. Moreover, the generation of circulating elastin-derived peptides reflects premature aging of the ECM. These extracellular elements contribute to the appearance of vascular rigidity, often the origin of pathologies such as hypertension and atherosclerosis. In conclusion, we show that diabetic mice aged 6 months present the same characteristics of ECM wear as those observed in mice aged 20 months. This accelerated aortic wall remodeling could then explain the early onset of cardiovascular diseases and, therefore, the premature death of DT2 patients.

2.
J Cell Physiol ; 239(6): e31257, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38504496

RESUMEN

Bone diseases are increasing with aging populations and it is important to identify clues to develop innovative treatments. Vasn, which encodes vasorin (Vasn), a transmembrane protein involved in the pathophysiology of several organs, is expressed during the development in intramembranous and endochondral ossification zones. Here, we studied the impact of Vasn deletion on the osteoblast and osteoclast dialog through a cell Coculture model. In addition, we explored the bone phenotype of Vasn KO mice, either constitutive or tamoxifen-inducible, or with an osteoclast-specific deletion. First, we show that both osteoblasts and osteoclasts express Vasn. Second, we report that, in both KO mouse models but not in osteoclast-targeted KO mice, Vasn deficiency was associated with an osteopenic bone phenotype, due to an imbalance in favor of osteoclastic resorption. Finally, through the Coculture experiments, we identify a dysregulation of the Wnt/ß-catenin pathway together with an increase in RANKL release by osteoblasts, which led to an enhanced osteoclast activity. This study unravels a direct role of Vasn in bone turnover, introducing a new biomarker or potential therapeutic target for bone pathologies.


Asunto(s)
Remodelación Ósea , Técnicas de Cocultivo , Osteoblastos , Osteoclastos , Vía de Señalización Wnt , Animales , Ratones , Huesos/metabolismo , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/patología , Remodelación Ósea/fisiología , Resorción Ósea/metabolismo , Resorción Ósea/genética , Resorción Ósea/patología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis/fisiología , Ligando RANK/metabolismo , Ligando RANK/genética
3.
J Cell Physiol ; 237(10): 3845-3859, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35892191

RESUMEN

Within the cardiovascular system, the protein vasorin (Vasn) is predominantly expressed by vascular smooth muscle cells (VSMCs) in the coronary arteries and the aorta. Vasn knockout (Vasn-/- ) mice die within 3 weeks of birth. In the present study, we investigated the role of vascular Vasn expression on vascular function. We used inducible Vasn knockout mice (VasnCRE-ERT KO and VasnSMMHC-CRE-ERT2 KO , in which respectively all cells or SMCs only are targeted) to analyze the consequences of total or selective Vasn loss on vascular function. Furthermore, in vivo effects were investigated in vitro using human VSMCs. The death of VasnCRE-ERT KO mice 21 days after tamoxifen injection was concomitant with decreases in blood pressure, angiotensin II levels, and vessel contractibility to phenylephrine. The VasnSMMHC-CRE-ERT2 KO mice displayed concomitant changes in vessel contractibility in response to phenylephrine and angiotensin II levels. In vitro, VASN deficiency was associated with a shift toward the SMC contractile phenotype, an increase in basal intracellular Ca2+ levels, and a decrease in the SMCs' ability to generate a calcium signal in response to carbachol or phenylephrine. Additionally, impaired endothelium-dependent relaxation (due to changes in nitric oxide signaling) was observed in all Vasn knockout mice models. Our present findings highlight the role played by Vasn SMC expression in the maintenance of vascular functions. The mechanistic experiments suggested that these effects are mediated by SMC phenotype switching and changes in intracellular calcium homeostasis, angiotensin II levels, and NO signaling.


Asunto(s)
Angiotensina II , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de la Membrana/metabolismo , Músculo Liso Vascular , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Calcio/metabolismo , Carbacol , Humanos , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Fenilefrina/metabolismo , Tamoxifeno
4.
Int J Mol Sci ; 23(11)2022 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-35682871

RESUMEN

The endothelium has a fundamental role in the cardiovascular complications of coronavirus disease 2019 (COVID-19). Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) particularly affects endothelial cells. The virus binds to the angiotensin-converting enzyme 2 (ACE-2) receptor (present on type 2 alveolar cells, bronchial epithelial cells, and endothelial cells), and induces a cytokine storm. The cytokines tumor necrosis factor alpha, interleukin-1 beta, and interleukin-6 have particular effects on endothelial cells-leading to endothelial dysfunction, endothelial cell death, changes in tight junctions, and vascular hyperpermeability. Under normal conditions, apoptotic endothelial cells are removed into the bloodstream. During COVID-19, however, endothelial cells are detached more rapidly, and do not regenerate as effectively as usual. The loss of the endothelium on the luminal surface abolishes all of the vascular responses mediated by the endothelium and nitric oxide production in particular, which results in greater contractility. Moreover, circulating endothelial cells infected with SARS-CoV-2 act as vectors for viral dissemination by forming clusters that migrate into the circulation and reach distant organs. The cell clusters and the endothelial dysfunction might contribute to the various thromboembolic pathologies observed in COVID-19 by inducing the formation of intravascular microthrombi, as well as by triggering disseminated intravascular coagulation. Here, we review the contributions of endotheliopathy and endothelial-cell-derived extracellular vesicles to the pathogenesis of COVID-19, and discuss therapeutic strategies that target the endothelium in patients with COVID-19.


Asunto(s)
COVID-19 , Enfermedades Vasculares , COVID-19/complicaciones , Citocinas/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , SARS-CoV-2 , Enfermedades Vasculares/metabolismo
5.
Kidney Int ; 99(6): 1382-1391, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33647324

RESUMEN

In chronic kidney disease (CKD), calcium-sensing receptor (CaSR) expression and function have been extensively studied in parathyroid tissue and vascular tissues. To examine whether similar changes occurred in other tissues, we measured total and surface CaSR expression in monocytes of patients with various stages of CKD and healthy volunteers respectively in cross-sectional studies. We further explored in vitro the impact of uremic serum on CaSR expression in monocytes (U937 and THP-1 cell lines), and whether human peripheral blood mononuclear cells or U937 and THP-1 monocytes might modify vascular calcium deposition in rat carotid arteries in vitro. CKD was associated with a decrease in peripheral blood mononuclear cell CaSR expression both in total and at the monocyte surface alone (43% and 34%, respectively in CKD stages 4-5). This decrease was associated with a reduction in the ability of monocytes to inhibit vascular calcification in vitro. Pretreatment with the calcimimetic NPSR568 of peripheral blood mononuclear cells isolated from patients with CKD significantly improved monocyte capacity to reduce carotid calcification in vitro. The fewer peripheral blood mononuclear cells expressing cell surface CaSR, the more calcimimetic treatment enhanced the decrease of carotid calcium content. Thus, we demonstrate that monocyte CaSR expression is decreased in patients with CKD and provide in vitro evidence for a potential role of this decrease in the promotion of vascular calcification. Hence, targeting this alteration or following monocyte CaSR expression as an accessible marker might represent a promising therapeutic strategy in CKD-associated arterial calcification.


Asunto(s)
Monocitos , Receptores Sensibles al Calcio , Insuficiencia Renal Crónica , Calcificación Vascular , Animales , Calcio , Estudios Transversales , Humanos , Leucocitos Mononucleares , Ratas , Calcificación Vascular/etiología , Calcificación Vascular/prevención & control
6.
Int J Mol Sci ; 21(12)2020 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-32560362

RESUMEN

This study addressed the hypothesis that soluble epoxide hydrolase (sEH), which metabolizes endothelium-derived epoxyeicosatrienoic acids, plays a role in vascular calcification. The sEH inhibitor trans-4-(4-(3-adamantan-1-yl-ureido)-cyclohexyloxy)-benzoic acid (t-AUCB) potentiated the increase in calcium deposition of rat aortic rings cultured in high-phosphate conditions. This was associated with increased tissue-nonspecific alkaline phosphatase activity and mRNA expression level of the osteochondrogenic marker Runx2. The procalcifying effect of t-AUCB was prevented by mechanical aortic deendothelialization or inhibition of the production and action of epoxyeicosatrienoic acids using the cytochrome P450 inhibitor fluconazole and the antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE), respectively. Similarly, exogenous epoxyeicosatrienoic acids potentiated the calcification of rat aortic rings through a protein kinase A (PKA)-dependent mechanism and of human aortic vascular smooth muscle cells when sEH was inhibited by t-AUCB. Finally, a global gene expression profiling analysis revealed that the mRNA expression level of sEH was decreased in human carotid calcified plaques compared to adjacent lesion-free sites and was inversely correlated with Runx2 expression. These results show that sEH hydrolase plays a protective role against vascular calcification by reducing the bioavailability of epoxyeicosatrienoic acids.


Asunto(s)
Susceptibilidad a Enfermedades , Epóxido Hidrolasas/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Metabolismo de los Lípidos , Calcificación Vascular/etiología , Calcificación Vascular/metabolismo , Animales , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Diferenciación Celular , Endotelio/metabolismo , Humanos , Monoéster Fosfórico Hidrolasas/metabolismo , ARN Mensajero/genética , Ratas
7.
Nephrol Dial Transplant ; 34(7): 1125-1134, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30481303

RESUMEN

BACKGROUND: Vascular calcification (VC) is amplified during chronic kidney disease, partly due to uraemic toxins such as inorganic phosphate (Pi) and indoxyl sulphate (IS) that trigger osteogenic differentiation of vascular smooth muscle cells (VSMCs). These toxins also alter endothelial cell (EC) functions but whether this contributes to VC is unknown. Here, we hypothesized that ECs exposed to Pi and IS promote VSMC calcification. METHODS: Human umbilical vein ECs were treated with Pi, IS or both, and then the conditioned media [endothelial cell conditioned medium (EC-CM)] was collected. Human aortic SMCs (HASMCs) were exposed to the same toxins, with or without EC-CM, and then calcification and osteogenic differentiation were evaluated. Procalcifying factors secreted from ECs in response to Pi and IS were screened. Rat aortic rings were isolated to assess Pi+IS-induced calcification at the tissue level. RESULTS: Pi and Pi+IS induced HASMCs calcification, which was significantly exacerbated by EC-CM. Pi+IS induced the expression and secretion of interleukin-8 (IL-8) from ECs. While IL-8 treatment of HASMCs stimulated the Pi+IS-induced calcification in a concentration-dependent manner, IL-8 neutralizing antibody, IL-8 receptors antagonist or silencing IL-8 gene expression in ECs before collecting EC-CM significantly prevented the EC-CM procalcifying effect. IL-8 did not promote the Pi+IS-induced osteogenic differentiation of HASMCs but prevented the induction of osteopontin (OPN), a potent calcification inhibitor. In rat aortic rings, IS also promoted Pi-induced calcification and stimulated the expression of IL-8 homologues. Interestingly, in the Pi+IS condition, IL-8 receptor antagonist lifted the inhibition of OPN expression and partially prevented aortic calcification. CONCLUSION: These results highlight a novel role of IL-8, whose contribution to VC in the uraemic state results at least from interaction between ECs and VSMCs.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Indicán/farmacología , Interleucina-8/metabolismo , Fosfatos/farmacología , Insuficiencia Renal Crónica/metabolismo , Calcificación Vascular/etiología , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Ratas , Ratas Wistar , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
8.
J Cell Physiol ; 230(12): 2927-35, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25899466

RESUMEN

Chronic kidney disease (CKD) is characterized by vascular remodeling and the retention of uremic toxins, several of which are independently associated with the high cardiovascular mortality rate in CKD patients. Whether the association between these uremic toxins and cardiovascular mortality is due to induction of vascular dysfunction and resulting vascular remodeling remains to be determined. This study evaluates the effects of para-cresyl sulfate (PCS), a newly identified uremic toxin, on vascular function and remodeling. PCS acutely induced oxidative stress in both endothelial and vascular smooth muscle cells, with a maximal effect at 0.15 mM, corresponding to the mean "uremic" concentration found in dialysis patients. PCS significantly increased within 30 min phenylephrine-induced contraction of mouse thoracic aorta, through direct activation of rho-kinase, independently of oxidative stress induction, as demonstrated by the capacity of rho-kinase inhibitor Y-27632 to abolish this effect. After exposure of the aorta to PCS for 48 h, we observed inward eutrophic remodeling, a hallmark of uremic vasculopathy characterized by a reduction of the area of both lumen and media, with unchanged media/lumen ratio. In conclusion, elevated PCS concentrations such as those observed in CKD patients, by promoting both vascular dysfunction and vascular remodeling, may contribute to the development of hypertension and to cardiovascular mortality in CKD.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Cresoles/toxicidad , Ésteres del Ácido Sulfúrico/toxicidad , Remodelación Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Animales , Aorta Torácica/metabolismo , Aorta Torácica/fisiopatología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Técnicas In Vitro , Ratones Endogámicos C57BL , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Quinasas Asociadas a rho/metabolismo
9.
Circ J ; 78(10): 2339-46, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25077548

RESUMEN

Chronic kidney disease (CKD) is characterized by high cardiovascular morbidity/mortality, which is linked in part to vascular calcification (VC) and endothelial dysfunction (ED). Hyperphosphatemia, a feature of CKD, is a well-known inducer of VC in preclinical models and is associated with poor outcomes in epidemiological studies. However, it remains to be seen whether lowering phosphate levels in CKD patients reduces VC and the morbidity/mortality rate. Furthermore, it is now clear from preclinical and clinical studies that phosphate is involved in ED. The present article reviews the direct and indirect mechanisms (eg, via fibroblast growth factor 23 and/or parathyroid hormone) by which hyperphosphatemia influence the onset of VC and ED in CKD.


Asunto(s)
Hiperfosfatemia , Fosfatos/sangre , Insuficiencia Renal Crónica , Calcificación Vascular , Animales , Endotelio Vascular/metabolismo , Humanos , Hiperfosfatemia/sangre , Hiperfosfatemia/etiología , Hiperfosfatemia/mortalidad , Hiperfosfatemia/terapia , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/mortalidad , Insuficiencia Renal Crónica/terapia , Calcificación Vascular/sangre , Calcificación Vascular/etiología , Calcificación Vascular/mortalidad , Calcificación Vascular/terapia
10.
Kidney Int ; 84(3): 491-500, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23594676

RESUMEN

Elevated serum phosphate and fibroblast growth factor 23 (FGF23) levels are associated with cardiovascular disease (CVD) in patients with chronic renal failure (CRF). The phosphate-binder sevelamer has been shown to decrease both phosphate and FGF23, but limited data indicate that sevelamer improves CRF-related CVD, such as diastolic dysfunction, left ventricular hypertrophy (LVH), and aortic stiffness. To gain additional information, we measured the effects of sevelamer on CVD in a murine model of CRF. Groups of CRF and sham-operated mice received regular chow or 3% sevelamer-HCl in the chow for 14 weeks, starting 6 weeks after the initiation of CRF or sham operation. After the first 8 weeks of sevelamer treatment, CRF mice had decreased serum phosphate levels and an improved aortic systolic expansion rate, pulse-wave velocity, and diastolic function, although LVH remained unchanged. Following an additional 6-week course of sevelamer, LVH had not progressed. The FGF23 serum level was not reduced by sevelamer until after 14 weeks of treatment. In multiple regression analysis, serum phosphate, but not FGF23, was independently correlated with LV diastolic function and mass. Thus, sevelamer first improved aortic stiffness and diastolic dysfunction and secondarily prevented LVH in mice with CRF. The phosphate-lowering, rather than FGF23-lowering, effect appears to be responsible for the observed cardiovascular improvement.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Fallo Renal Crónico/tratamiento farmacológico , Poliaminas/farmacología , Poliaminas/uso terapéutico , Rigidez Vascular/efectos de los fármacos , Animales , Sistema Cardiovascular/fisiopatología , Quelantes/farmacología , Quelantes/uso terapéutico , Diástole/efectos de los fármacos , Diástole/fisiología , Modelos Animales de Enfermedad , Femenino , Factor-23 de Crecimiento de Fibroblastos , Hipertrofia Ventricular Izquierda/fisiopatología , Riñón/metabolismo , Fallo Renal Crónico/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosfatos/metabolismo , Análisis de la Onda del Pulso , Análisis de Regresión , Sevelamer , Rigidez Vascular/fisiología
11.
Front Med (Lausanne) ; 10: 1186122, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37332749

RESUMEN

Objectives: To evaluate extracellular vesicles levels in a cohort of SARS-CoV-2's patients hospitalized in an intensive care unit with and without COVID-19 associated thromboembolic events. Methods: In this study, we aim to assess endothelial and platelet membrane-derived extracellular vesicles levels in a cohort of SARS-CoV-2 patients with and without COVID-19-associated thromboembolic events who were hospitalized in an intensive care unit. Annexin-V positive extracellular vesicles levels were prospectively assessed by flow cytometry in one hundred twenty-three critically ill adults diagnosed with acute respiratory distress syndrome associated with a SARS-CoV-2 infection, ten adults diagnosed for moderate SARS-CoV-2 infection and 25 healthy volunteers. Results: On our critically ill patients, thirty-four patients (27.6%) had a thromboembolic event, Fifty-three (43%) died. Endothelial and platelet membrane-derived extracellular vesicles were drastically increased in SARS-CoV-2 patients hospitalized in the ICU compared to healthy volunteers. Moreover a slighty higher small/large ratio for platelets membrane-derived extracellular vesicles in patients was linked to thrombo-embolic events. Conclusion: A comparison between total annexin-V positive extracellular vesicles levels in severe and moderate SARS-CoV-2 infection and healthy controls showed a significant increase in patients with severe infection and their sizes could be considered as biomarkers of SARS-CoV-2 associated thrombo-embolic events.

12.
Toxins (Basel) ; 14(5)2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35622583

RESUMEN

Acute kidney injury (AKI) is a frequent disease encountered in the hospital, with a higher incidence in intensive care units. Despite progress in renal replacement therapy, AKI is still associated with early and late complications, especially cardiovascular events and mortality. The role of gut-derived protein-bound uremic toxins (PBUTs) in vascular and cardiac dysfunction has been extensively studied during chronic kidney disease (CKD), in particular, that of indoxyl sulfate (IS), para-cresyl sulfate (PCS), and indole-3-acetic acid (IAA), resulting in both experimental and clinical evidence. PBUTs, which accumulate when the excretory function of the kidneys is impaired, have a deleterious effect on and cause damage to cardiovascular tissues. However, the link between PBUTs and the cardiovascular complications of AKI and the pathophysiological mechanisms potentially involved are unclear. This review aims to summarize available data concerning the participation of PBUTs in the early and late cardiovascular complications of AKI.


Asunto(s)
Lesión Renal Aguda , Cardiopatías , Insuficiencia Renal Crónica , Lesión Renal Aguda/etiología , Humanos , Riñón , Insuficiencia Renal Crónica/complicaciones , Tóxinas Urémicas
13.
Circulation ; 119(2): 306-13, 2009 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-19118252

RESUMEN

BACKGROUND: Chronic renal failure (CRF) is associated with cardiac dysfunction and increased aortic stiffness. The mechanisms involved are not clearly understood. We examined changes over time in cardiac and aortic function in a murine CRF model. METHODS AND RESULTS: Eight-week-old mice were randomly assigned to 1 of 4 groups: wild-type non-CRF, wild-type CRF, apolipoprotein E knockout non-CRF, and apolipoprotein E knockout CRF. Echocardiography was performed and blood samples were taken at baseline and after 6 and 10 weeks of CRF. Vascular reactivity and adhesion molecule expression were studied after 6 and 10 weeks of CRF. Left ventricular hypertrophy, altered left ventricular relaxation, and increased aortic stiffness were observed after 6 weeks of CRF and persisted after 10 weeks. The 4 groups of mice did not significantly differ in terms of arterial blood pressure and aortic structure. The degree of vascular calcification and serum total cholesterol concentration were higher in the CRF groups than in the non-CRF groups. These changes, however, could not explain the cardiac and vascular differences seen in the 2 CRF groups. In contrast, alterations in vascular reactivity, the upregulation of adhesion molecule expression, and CRF status were significantly associated with these changes. CONCLUSIONS: In a mouse model of CRF, left ventricular hypertrophy, cardiac diastolic dysfunction, and increased aortic stiffness were not related to structural changes in the aorta (including aortic calcification) or high serum cholesterol levels. However, cardiac and aortic abnormalities were associated with the extent of subendothelial dysfunction and the severity of CRF.


Asunto(s)
Enfermedades de la Aorta/fisiopatología , Calcinosis/fisiopatología , Fallo Renal Crónico/fisiopatología , Uremia/fisiopatología , Animales , Aorta , Enfermedades de la Aorta/sangre , Calcinosis/sangre , Cardiomiopatías/sangre , Cardiomiopatías/fisiopatología , Colesterol/sangre , Modelos Animales de Enfermedad , Femenino , Hipertrofia Ventricular Izquierda/sangre , Hipertrofia Ventricular Izquierda/fisiopatología , Técnicas In Vitro , Fallo Renal Crónico/sangre , Fallo Renal Crónico/diagnóstico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria , Uremia/sangre
14.
Toxins (Basel) ; 12(6)2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32570781

RESUMEN

Vascular dysfunction is an essential element found in many cardiovascular pathologies and in pathologies that have a cardiovascular impact such as chronic kidney disease (CKD). Alteration of vasomotricity is due to an imbalance between the production of relaxing and contracting factors. In addition to becoming a determining factor in pathophysiological alterations, vascular dysfunction constitutes the first step in the development of atherosclerosis plaques or vascular calcifications. In patients with CKD, alteration of vasomotricity tends to emerge as being a new, less conventional, risk factor. CKD is characterized by the accumulation of uremic toxins (UTs) such as phosphate, para-cresyl sulfate, indoxyl sulfate, and FGF23 and, consequently, the deleterious role of UTs on vascular dysfunction has been explored. This accumulation of UTs is associated with systemic alterations including inflammation, oxidative stress, and the decrease of nitric oxide production. The present review proposes to summarize our current knowledge of the mechanisms by which UTs induce vascular dysfunction.


Asunto(s)
Vasos Sanguíneos/metabolismo , Cresoles/sangre , Indicán/sangre , Insuficiencia Renal Crónica/complicaciones , Ésteres del Ácido Sulfúrico/sangre , Uremia/complicaciones , Enfermedades Vasculares/etiología , Animales , Vasos Sanguíneos/fisiopatología , Factor-23 de Crecimiento de Fibroblastos , Humanos , Mediadores de Inflamación/metabolismo , Estrés Oxidativo , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/terapia , Uremia/sangre , Uremia/fisiopatología , Uremia/terapia , Enfermedades Vasculares/sangre , Enfermedades Vasculares/fisiopatología , Enfermedades Vasculares/prevención & control
15.
Curr Vasc Pharmacol ; 18(5): 507-516, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31284864

RESUMEN

OBJECTIVE: Restenosis is a frequent complication of angioplasty. It consists of a neointimal hyperplasia resulting from progression and migration of vascular smooth muscle cells (VSMC) into the vessel lumen. microRNA miR-223 has recently been shown to be involved in cardiovascular diseases including atherosclerosis, vascular calcification and arterial thrombosis. In this study, our aim was to assess the impact of miR-223 modulation on restenosis in a rat model of carotid artery after balloon injury. METHODS: The over and down-expression of miR-223 was induced by adenoviral vectors, containing either a pre-miR-223 sequence allowing artificial miR-223 expression or a sponge sequence, trapping the native microRNA, respectively. Restenosis was quantified on stained rat carotid sections. RESULTS: In vitro, three mRNA (Myocyte Enhancer Factor 2C (MEF2C), Ras homolog gene family, member B (RhoB) and Nuclear factor 1 A-type (NFIA)) reported as miR-223 direct targets and known to be implicated in VSMC differentiation and contractility were studied by RT-qPCR. Our findings showed that down-expression of miR-223 significantly reduced neointimal hyperplasia by 44% in carotids, and was associated with a 2-3-fold overexpression of MEF2C, RhoB and NFIA in a murine monocyte macrophage cell line, RAW 264.7 cells. CONCLUSION: Down-regulating miR-223 could be a potential therapeutic approach to prevent restenosis after angioplasty.


Asunto(s)
Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/terapia , Estenosis Carotídea/prevención & control , Terapia Genética , MicroARNs/metabolismo , Angioplastia de Balón , Animales , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Estenosis Carotídea/genética , Estenosis Carotídea/metabolismo , Estenosis Carotídea/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Masculino , Ratones , MicroARNs/genética , Neointima , Células RAW 264.7 , Ratas Wistar , Transducción de Señal
16.
Europace ; 10(1): 79-85, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18156160

RESUMEN

AIMS: The distinct cardiac arrhythmia diseases, Brugada syndrome (BS) and isolated cardiac conduction disease (ICCD) are caused by heterozygous mutations in the SCN5A gene. Previous studies have demonstrated an intriguing association between ICCD and BS with the same mutation in the SCN5A gene. METHODS AND RESULTS: The proband of a multigenerational family presented BS and a familial history of sudden death. We performed clinical evaluations in family members including drug testing and screening for SCN5A mutations. Based on electrocardiogram features, we identified four individuals with BS, two with ICCD and one compatible with both. For five individuals, one with BS and ICCD, three with BS and one with ICCD, we characterized a heterozygous C- to T- mutation at position 4313 (P1438L) in the SCN5A gene. Expression studies of the P1438L mutation showed non-functional channels. The proband's father with the BS phenotype was not a carrier of the new SCN5A mutation. CONCLUSION: We report the case of a family with BS and/or ICCD and describe a novel mutation, the P1438L SCN5A mutation. In this family, the occurrence of BS and ICCD could be due to this single mutation but also to the accidental association of both diseases.


Asunto(s)
Arritmias Cardíacas/genética , Síndrome de Brugada/genética , Sistema de Conducción Cardíaco/fisiopatología , Proteínas Musculares/genética , Mutación/genética , Canales de Sodio/genética , Adulto , Anciano , Arritmias Cardíacas/fisiopatología , Síndrome de Brugada/fisiopatología , Electrocardiografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Canal de Sodio Activado por Voltaje NAV1.5 , Linaje , Polimorfismo de Nucleótido Simple
17.
N Engl J Med ; 349(4): 343-9, 2003 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-12878741

RESUMEN

BACKGROUND: Aortic-valve stenosis can be complicated by bleeding that is associated with acquired type 2A von Willebrand syndrome. However, the prevalence and cause of the hemostatic abnormality in aortic stenosis are unknown. METHODS: We enrolled 50 consecutive patients with aortic stenosis, who completed a standardized screening questionnaire to detect a history of bleeding. Forty-two patients with severe aortic stenosis underwent valve replacement. Platelet function under conditions of high shear stress, von Willebrand factor collagen-binding activity and antigen levels, and the multimeric structure of von Willebrand factor were assessed at base line and one day, seven days, and six months postoperatively. RESULTS: Skin or mucosal bleeding occurred in 21 percent of the patients with severe aortic stenosis. Platelet-function abnormalities under conditions of high shear stress, decreased von Willebrand factor collagen-binding activity and the loss of the largest multimers, or a combination of these was present in 67 to 92 percent of patients with severe aortic stenosis and correlated significantly with the severity of valve stenosis. Primary hemostatic abnormalities were completely corrected on the first day after surgery but tended to recur at six months, especially when there was a mismatch between patient and prosthesis (with an effective orifice area of less than 0.8 cm2 per square meter of body-surface area). CONCLUSIONS: Type 2A von Willebrand syndrome is common in patients with severe aortic stenosis. Von Willebrand factor abnormalities are directly related to the severity of aortic stenosis and are improved by valve replacement in the absence of mismatch between patient and prosthesis.


Asunto(s)
Estenosis de la Válvula Aórtica/complicaciones , Hemorragia/etiología , Enfermedades de von Willebrand/etiología , Factor de von Willebrand/metabolismo , Anciano , Estenosis de la Válvula Aórtica/sangre , Estenosis de la Válvula Aórtica/cirugía , Femenino , Estudios de Seguimiento , Prótesis Valvulares Cardíacas , Implantación de Prótesis de Válvulas Cardíacas , Hemorreología , Humanos , Masculino , Persona de Mediana Edad , Hemorragia Posoperatoria , Prevalencia , Encuestas y Cuestionarios , Factor de von Willebrand/química
18.
Joint Bone Spine ; 84(2): 175-181, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27346252

RESUMEN

OBJECTIVES: We assessed calcium-sensing receptor (CaSR) expression in monocytes isolated from synovial fluid of patients with different types of rheumatisms and explored whether CaSR expression was related to the inflammatory nature of synovial fluid. METHODS: Forty-one patients were included: osteoarthritis (n=10), microcristallin rheumatisms (n=10), rheumatoid arthritis (n=12) and other inflammatory rheumatisms (n=9). Surface and total CaSR expressions in monocytes isolated from synovial fluid and blood were assessed by flow cytometry analysis. U937 cells were cultured during 24hours in presence of cell-free synovial fluids. RESULTS: Every monocyte population tested express the CaSR intra- and extracellularly. Whereas similar pattern of CaSR expression exist in monocyte isolated from blood or synovial fluids, our results indicate that higher CaSR expression levels can be observed in monocytes from synovial fluids than in circulating monocytes. In both populations of monocytes, surface and total CaSR expressions were found to be significantly increased in patients with osteoarthritis compared to rheumatoid arthritis. Similar data were obtained when U937 cells were incubated with cell-free synovial fluids from osteoarthritis patients. Still present, this effect was significantly lowered when "inflammatory" synovial fluids were introduced in culture. CONCLUSIONS: Our results indicate that CaSR expression in synovial derived monocytes is higher in osteoarthritis than in inflammatory rheumatisms and that CaSR expression is modulated by the nature of the synovial fluid. Given the role played by monocytes in the pathogenesis of chronic rheumatisms, monocytes could be interesting therapeutic targets via the CaSR.


Asunto(s)
Artritis/metabolismo , Monocitos/metabolismo , Receptores Sensibles al Calcio/biosíntesis , Líquido Sinovial/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Estudios Transversales , Femenino , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , Osteoartritis/metabolismo , Proyectos Piloto , Células U937 , Adulto Joven
19.
J Tissue Eng Regen Med ; 11(2): 382-389, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-24919776

RESUMEN

In 2000, Masquelet reported a long bone reconstruction technique using an induced membrane formed around a polymethylmethacrylate (PMMA) spacer placed in the defect with appropriate stabilization followed by secondary bone graft after PMMA removal. This reconstruction procedure allows rapid and safe bone reformation for septic, traumatic, neoplastic or congenital bone defects. A rat model of the Masquelet technique was developed to further characterize the biological activities of this induced membrane. Our model allows healing of a critical-sized femoral defect (8 mm) by means of this procedure over a period of 18 weeks. Comparison of induced membranes obtained 3, 4, 5 and 6 weeks after PMMA insertion indicated that this tissue changes over time. Several mineralization spots and bone cells were observed in contact with the PMMA, when assessed by Alizarin Red, Von Kossa, Alkaline phosphatase and Tartrate-resistant acid phosphatase staining of the membranes. CTR (calcitonin receptor)- and RANK (Receptor Activator of Nuclear factor Kappa B)- positive mononuclear cells were detected in the induced membrane, confirming the presence of osteoclasts in this tissue. These cells were observed in a thin, highly cellular layer in the induced membrane in contact with the PMMA. Together, these findings suggest that the membrane is able to promote osteointegration of autologous corticocancellous bone grafts during the Masquelet technique by creating local conditions that may be favourable to graft bone remodelling and osteointegration. Copyright © 2014 John Wiley & Sons, Ltd.


Asunto(s)
Regeneración Ósea , Huesos/patología , Membranas Artificiales , Osteoclastos/citología , Procedimientos de Cirugía Plástica , Animales , Cementos para Huesos , Trasplante Óseo , Fémur/patología , Leucocitos Mononucleares/metabolismo , Masculino , Polimetil Metacrilato/química , Ratas , Ratas Sprague-Dawley , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Receptores de Calcitonina/metabolismo , Procedimientos Quirúrgicos Operativos , Factores de Tiempo , Trasplante Autólogo , Cicatrización de Heridas
20.
Curr Vasc Pharmacol ; 15(1): 40-46, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27633456

RESUMEN

The discovery of small RNAs has shed new light on microRNA (mRNA) regulation and a range of biological processes. The recognition that miRNAs-221 and -222 are sensitive regulators in the endothelium may enable the identification of novel biomarkers and therapeutic targets. Given that endothelial dysfunction precedes the development of atherosclerosis and contributes to the development of cardiovascular damage, circulating miRNAs produced by Endothelial Cells (ECs) are putative biomarkers for a wide range of cardiovascular diseases. Furthermore, EC proliferation and migration have a critical role in the formation of new blood vessels (angiogenesis), an important component of physiological processes and tumour growth. Hence, the use of anti-angiogenic miRNAs might constitute a novel therapeutic strategy. Along with endothelial angiogenesis, several other processes involving ECs (such as neointimal lesion formation, vascular inflammation, lipoprotein metabolism and hypertension) are critical factors in atherogenesis and atherosclerosis. The fact that human blood-derived progenitor cells, endothelial progenitor cells, umbilical vein ECs and quiescent ECs express high levels of miR-221/222 suggests an important role for this miRNA cluster in endothelial (patho) physiology. The present article reviews current knowledge on miRNAs-221/222's regulation roles in endothelial function and disease in general and angiogenesis in particular.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Células Endoteliales/metabolismo , MicroARNs/metabolismo , Neovascularización Fisiológica , Animales , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/fisiopatología , Proliferación Celular , Senescencia Celular , Células Endoteliales/patología , Regulación de la Expresión Génica , Humanos , MicroARNs/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA