Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 132(8): 993-1012, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37053279

RESUMEN

Patients with chronic kidney disease (CKD) exhibit tremendously elevated risk for cardiovascular disease, particularly ischemic heart disease, due to premature vascular and cardiac aging and accelerated ectopic calcification. The presence of cardiovascular calcification associates with increased risk in patients with CKD. Disturbed mineral homeostasis and diverse comorbidities in these patients drive increased systemic cardiovascular calcification in different manifestations with diverse clinical consequences, like plaque instability, vessel stiffening, and aortic stenosis. This review outlines the heterogeneity in calcification patterning, including mineral type and location and potential implications on clinical outcomes. The advent of therapeutics currently in clinical trials may reduce CKD-associated morbidity. Development of therapeutics for cardiovascular calcification begins with the premise that less mineral is better. While restoring diseased tissues to a noncalcified homeostasis remains the ultimate goal, in some cases, calcific mineral may play a protective role, such as in atherosclerotic plaques. Therefore, developing treatments for ectopic calcification may require a nuanced approach that considers individual patient risk factors. Here, we discuss the most common cardiac and vascular calcification pathologies observed in CKD, how mineral in these tissues affects function, and the potential outcomes and considerations for therapeutic strategies that seek to disrupt the nucleation and growth of mineral. Finally, we discuss future patient-specific considerations for treating cardiac and vascular calcification in patients with CKD-a population in need of anticalcification therapies.


Asunto(s)
Enfermedades Cardiovasculares , Insuficiencia Renal Crónica , Calcificación Vascular , Humanos , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/etiología , Enfermedades Cardiovasculares/etiología , Minerales , Envejecimiento
2.
Arterioscler Thromb Vasc Biol ; 43(8): 1429-1440, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37381986

RESUMEN

BACKGROUND: Increasing evidence suggests that superoxide ions produced by NOX (nicotinamide adenine dinucleotide phosphate oxidases) mediate vascular effects of Ang II (angiotensin II) evoked by atherogenic diets. Here, we analyzed the mechanism by which NOX2 contributes to Ang II-induced ET-1 (endothelin 1) production in human microvascular endothelial cells. METHODS: The effects of high-fat diet were compared between WT (wild type) and Nox2 (mouse NOX2 gene)-deficient mice. ET-1 production and NOX2 expression by human microvascular endothelial cells in vitro were analyzed by ELISA, reverse transcription quantitative polymerase chain reaction, electrophoretic mobility shift assay, promoter deletions, RNA interference, and pharmacological inhibition. Production of superoxide anions was visualized by fluorescent cell labeling. RESULTS: Feeding mice high-fat diet for 10 weeks increased cardiac expression and plasma levels of Ang II and ET-1 in WT but not in Nox2-deficient animals. Exposure of human microvascular endothelial cells to Ang II resulted in increased ET-1 production, which could be blocked by silencing NOX2 (human NOX2 gene). Ang II promoted NOX2 expression through induction of the Oct-1 (human/mouse octamer binding transcription factor 1 protein) and activation of the NOX2 promoter region containing Oct-1-binding sites. Stimulation of NOX2 expression by Ang II was associated with increased production of superoxide anions. Inhibition of Oct-1 by small interfering RNA reduced Ang II-induced NOX2 expression and superoxide anion production, and neutralization of superoxide by SOD (superoxide dismutase) abolished Ang II-stimulated ET1 (human ET-1 gene) promoter activity, ET1 mRNA expression, and ET-1 release. CONCLUSIONS: Ang II may promote ET-1 production in the endothelium in response to atherogenic diets through a mechanism that involves the transcription factor Oct-1 and the increased formation of superoxide anions by NOX2.


Asunto(s)
Células Endoteliales , Superóxidos , Ratones , Animales , Humanos , Superóxidos/metabolismo , Células Endoteliales/metabolismo , Factor 1 de Transcripción de Unión a Octámeros , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Angiotensina II/farmacología , Angiotensina II/metabolismo , Especies Reactivas de Oxígeno/metabolismo
3.
Eur Heart J ; 44(10): 885-898, 2023 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-36660854

RESUMEN

AIMS: Calcific aortic valve disease (CAVD) is the most common valve disease, which consists of a chronic interplay of inflammation, fibrosis, and calcification. In this study, sortilin (SORT1) was identified as a novel key player in the pathophysiology of CAVD, and its role in the transformation of valvular interstitial cells (VICs) into pathological phenotypes is explored. METHODS AND RESULTS: An aortic valve (AV) wire injury (AVWI) mouse model with sortilin deficiency was used to determine the effects of sortilin on AV stenosis, fibrosis, and calcification. In vitro experiments employed human primary VICs cultured in osteogenic conditions for 7, 14, and 21 days; and processed for imaging, proteomics, and transcriptomics including single-cell RNA-sequencing (scRNA-seq). The AVWI mouse model showed reduced AV fibrosis, calcification, and stenosis in sortilin-deficient mice vs. littermate controls. Protein studies identified the transition of human VICs into a myofibroblast-like phenotype mediated by sortilin. Sortilin loss-of-function decreased in vitro VIC calcification. ScRNA-seq identified 12 differentially expressed cell clusters in human VIC samples, where a novel combined inflammatory myofibroblastic-osteogenic VIC (IMO-VIC) phenotype was detected with increased expression of SORT1, COL1A1, WNT5A, IL-6, and serum amyloid A1. VICs sequenced with sortilin deficiency showed decreased IMO-VIC phenotype. CONCLUSION: Sortilin promotes CAVD by mediating valvular fibrosis and calcification, and a newly identified phenotype (IMO-VIC). This is the first study to examine the role of sortilin in valvular calcification and it may render it a therapeutic target to inhibit IMO-VIC emergence by simultaneously reducing inflammation, fibrosis, and calcification, the three key pathological processes underlying CAVD.


Asunto(s)
Estenosis de la Válvula Aórtica , Calcinosis , Humanos , Animales , Ratones , Estenosis de la Válvula Aórtica/genética , Válvula Aórtica/patología , Calcinosis/metabolismo , Constricción Patológica , Células Cultivadas , Fibrosis
4.
Int J Mol Sci ; 24(3)2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36768960

RESUMEN

Mitochondria are dynamic organelles regulating metabolism, cell death, and energy production. Therefore, maintaining mitochondrial health is critical for cellular homeostasis. Mitophagy and mitochondrial reorganization via fission and fusion are established mechanisms for ensuring mitochondrial quality. In recent years, mitochondrial-derived vesicles (MDVs) have emerged as a novel cellular response. MDVs are shed from the mitochondrial surface and can be directed to lysosomes or peroxisomes for intracellular degradation. MDVs may contribute to cardiovascular disease (CVD) which is characterized by mitochondrial dysfunction. In addition, evidence suggests that mitochondrial content is present in extracellular vesicles (EVs). Herein, we provide an overview of the current knowledge on MDV formation and trafficking. Moreover, we review recent findings linking MDV and EV biogenesis and discuss their role in CVD. Finally, we discuss the role of vesicle-mediated mitochondrial transfer and its potential cardioprotective effects.


Asunto(s)
Enfermedades Cardiovasculares , Vesículas Extracelulares , Humanos , Enfermedades Cardiovasculares/metabolismo , Mitocondrias/metabolismo , Lisosomas/metabolismo , Peroxisomas/metabolismo , Vesículas Extracelulares/metabolismo
5.
Int J Mol Sci ; 24(15)2023 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-37569721

RESUMEN

Osteoarthritis (OA) is a chronic joint disease characterized by articular cartilage calcification, loss of articular cartilage, bone changes, pain, and disability. Cartilage calcification is one hallmark of OA and is predominantly caused by basic calcium crystals formed due to an imbalance of the pyrophosphate pathway. Sortilin is a transmembrane protein that contributes to vascular calcification in atherosclerosis by externalizing alkaline phosphatase (ALP)-containing vesicles. Calcification in atherosclerosis and osteoarthritis has been associated with cellular senescence. The aim of this study was to investigate the potential role of sortilin and senescence in osteoarthritis-dependent cartilage calcification. Osteoarthritic cartilage from human knee joints was collected after joint replacement, and samples were analyzed by immunohistochemistry and quantitative RT-PCR analysis. Human chondrocytes were treated with osteogenic medium for up to 21 days to induce calcification. Western blots for sortilin and ALP, as well as an ALP activity assay, were performed. Human chondrocytes were treated with mitomycin C to induce senescence, and sortilin expression was quantified at the protein and gene levels. Sections of knee joints from a murine model of osteoarthritis were stained for sortilin and p16 and analyzed by immunohistochemistry. Treatment of wild-type chondrocytes using an osteogenic medium similar to human chondrocytes was performed. Osteoarthritic cartilage from mouse and human knee joints showed an increased number of sortilin and p16-positive chondrocytes compared to healthy cartilage. This observation was corroborated by increased gene expression of sortilin and p16 in mild and moderate osteoarthritic cartilage samples. To investigate the mechanism of sortilin regulation, human chondrocytes were treated with osteogenic medium to induce calcification. Sortilin protein levels and expression were increased after 7 days of stimulation, whereas ALP levels and activity were upregulated after 21 days of stimulation. Similar observations were made in a murine osteoarthritis model. Mechanistically, senescent chondrocytes induced by mitomycin C showed an upregulation of sortilin and ALP gene expression compared to non-senescent chondrocytes. Our data indicate that sortilin and ALP are upregulated during cartilage calcification, which is associated with chondrocyte senescence and thus might contribute to the pathogenesis of osteoarthritis. Cellular senescence seems to induce sortilin expression.

6.
Kidney Int ; 101(3): 574-584, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34767831

RESUMEN

Sortilin, an intracellular sorting receptor, has been identified as a cardiovascular risk factor in the general population. Patients with chronic kidney disease (CKD) are highly susceptible to develop cardiovascular complications such as calcification. However, specific CKD-induced posttranslational protein modifications of sortilin and their link to cardiovascular calcification remain unknown. To investigate this, we examined two independent CKD cohorts for carbamylation of circulating sortilin and detected increased carbamylated sortilin lysine residues in the extracellular domain of sortilin with kidney function decline using targeted mass spectrometry. Structure analysis predicted altered ligand binding by carbamylated sortilin, which was verified by binding studies using surface plasmon resonance measurement, showing an increased affinity of interleukin 6 to in vitro carbamylated sortilin. Further, carbamylated sortilin increased vascular calcification in vitro and ex vivo that was accelerated by interleukin 6. Imaging by mass spectrometry of human calcified arteries revealed in situ carbamylated sortilin. In patients with CKD, sortilin carbamylation was associated with coronary artery calcification, independent of age and kidney function. Moreover, patients with carbamylated sortilin displayed significantly faster progression of coronary artery calcification than patients without sortilin carbamylation. Thus, carbamylated sortilin may be a risk factor for cardiovascular calcification and may contribute to elevated cardiovascular complications in patients with CKD.


Asunto(s)
Insuficiencia Renal Crónica , Calcificación Vascular , Proteínas Adaptadoras del Transporte Vesicular , Humanos , Carbamilación de Proteína , Procesamiento Proteico-Postraduccional , Calcificación Vascular/etiología
7.
J Biol Chem ; 295(36): 12605-12617, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32647007

RESUMEN

In the heart, the serine carboxypeptidase cathepsin A (CatA) is distributed between lysosomes and the extracellular matrix (ECM). CatA-mediated degradation of extracellular peptides may contribute to ECM remodeling and left ventricular (LV) dysfunction. Here, we aimed to evaluate the effects of CatA overexpression on LV remodeling. A proteomic analysis of the secretome of adult mouse cardiac fibroblasts upon digestion by CatA identified the extracellular antioxidant enzyme superoxide dismutase (EC-SOD) as a novel substrate of CatA, which decreased EC-SOD abundance 5-fold. In vitro, both cardiomyocytes and cardiac fibroblasts expressed and secreted CatA protein, and only cardiac fibroblasts expressed and secreted EC-SOD protein. Cardiomyocyte-specific CatA overexpression and increased CatA activity in the LV of transgenic mice (CatA-TG) reduced EC-SOD protein levels by 43%. Loss of EC-SOD-mediated antioxidative activity resulted in significant accumulation of superoxide radicals (WT, 4.54 µmol/mg tissue/min; CatA-TG, 8.62 µmol/mg tissue/min), increased inflammation, myocyte hypertrophy (WT, 19.8 µm; CatA-TG, 21.9 µm), cellular apoptosis, and elevated mRNA expression of hypertrophy-related and profibrotic marker genes, without affecting intracellular detoxifying proteins. In CatA-TG mice, LV interstitial fibrosis formation was enhanced by 19%, and the type I/type III collagen ratio was shifted toward higher abundance of collagen I fibers. Cardiac remodeling in CatA-TG was accompanied by an increased LV weight/body weight ratio and LV end diastolic volume (WT, 50.8 µl; CatA-TG, 61.9 µl). In conclusion, CatA-mediated EC-SOD reduction in the heart contributes to increased oxidative stress, myocyte hypertrophy, ECM remodeling, and inflammation, implicating CatA as a potential therapeutic target to prevent ventricular remodeling.


Asunto(s)
Catepsina A/metabolismo , Miocitos Cardíacos/metabolismo , Proteolisis , Superóxido Dismutasa/metabolismo , Remodelación Ventricular , Animales , Catepsina A/genética , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología , Superóxido Dismutasa/genética
8.
Basic Res Cardiol ; 116(1): 57, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34647168

RESUMEN

The adrenal glands participate in cardiovascular (CV) physiology and the pathophysiology of CV diseases through their effects on sodium and water metabolism, vascular tone and cardiac function. In the present study, we identified a new adrenal compound controlling mesenchymal cell differentiation that regulates osteoblastic differentiation in the context of vascular calcification. This peptide was named the "calcification blocking factor" (CBF) due to its protective effect against vascular calcification and is released from chromogranin A via enzymatic cleavage by calpain 1 and kallikrein. CBF reduced the calcium content of cells and thoracic aortic rings under calcifying culture conditions, as well as in aortas from animals treated with vitamin D and nicotine (VDN animals). Furthermore, CBF prevented vascular smooth muscle cell (VSMC) transdifferentiation into osteoblast-like cells within the vascular wall via the sodium-dependent phosphate transporter PIT-1 and by inhibition of NF-κB activation and the subsequent BMP2/p-SMAD pathway. Pulse pressure, a marker of arterial stiffness, was significantly decreased in VDN animals treated with CBF. In line with our preclinical data, CBF concentration is significantly reduced in diseases characterized by increased calcification, as shown in patients with chronic kidney disease. In preparation for clinical translation, the active site of the native 19-AS long native CBF was identified as EGQEEEED. In conclusion, we have identified the new peptide CBF, which is secreted from the adrenal glands and might prevent vascular calcification by inhibition of osteogenic transdifferentiation. The anti-calcific effects of CBF and short active site may therefore promote the development of new tools for the prevention and/or treatment of vascular calcification.


Asunto(s)
Transdiferenciación Celular , Calcificación Vascular , Animales , Células Cultivadas , Cromogranina A , Humanos , Músculo Liso Vascular , Miocitos del Músculo Liso , Calcificación Vascular/prevención & control
9.
Nephrol Dial Transplant ; 35(1): 65-73, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30715488

RESUMEN

BACKGROUND: Optimal phosphate control is an unmet need in chronic kidney disease (CKD). High serum phosphate increases calcification burden and is associated with mortality and cardiovascular disease in CKD. Nicotinamide (NA) alone or in combination with calcium-free phosphate binders might be a strategy to reduce phosphate levels and calcification and thus impact cardiovascular disease in CKD. METHODS: We studied the effect of NA alone and in combination with magnesium carbonate (MgCO3) as a potential novel treatment strategy. CKD was induced in dilute brown non-agouti/2 mice by subtotal nephrectomy followed by a high-phosphate diet (HP) and 7 weeks of treatment with NA, MgCO3 or their combination. Control mice underwent subtotal nephrectomy and received an HP or underwent sham surgery and received standard chow plus NA. RESULTS: CKD mice showed increased serum fibroblast growth factor 23 and calcium-phosphate product that was normalized by all treatment regimes. NA alone increased soft tissue and vascular calcification, whereas any treatment with MgCO3 significantly reduced calcification severity in CKD. While MgCO3 supplementation alone resulted in decreased calcification severity, it resulted in increased intestinal expression of the phosphate transporters type II sodium-dependent phosphate transporter 1 (Pit-1). Combined therapy of MgCO3 and NA reduced tissue calcification and normalized expression levels of intestinal phosphate transporter proteins. CONCLUSIONS: In conclusion, the data indicate that NA increases while MgCO3 reduces ectopic calcification severity. Augmented expression of intestinal phosphate transporters by MgCO3 treatment was abolished by the addition of NA. However, the clinical relevance of the latter remains to be explored. Importantly, the data suggest no benefit of NA regarding treatment of calcification in addition to MgCO3.


Asunto(s)
Magnesio/farmacología , Músculo Liso Vascular/efectos de los fármacos , Niacinamida/farmacología , Insuficiencia Renal Crónica/complicaciones , Uremia/complicaciones , Calcificación Vascular/prevención & control , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos DBA , Músculo Liso Vascular/citología , Calcificación Vascular/etiología , Complejo Vitamínico B/farmacología
10.
Circ Res ; 121(3): 220-233, 2017 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-28607103

RESUMEN

RATIONALE: Mitochondrial changes occur during cell differentiation and cardiovascular disease. DRP1 (dynamin-related protein 1) is a key regulator of mitochondrial fission. We hypothesized that DRP1 plays a role in cardiovascular calcification, a process involving cell differentiation and a major clinical problem with high unmet needs. OBJECTIVE: To examine the effects of osteogenic promoting conditions on DRP1 and whether DRP1 inhibition alters the development of cardiovascular calcification. METHODS AND RESULTS: DRP1 was enriched in calcified regions of human carotid arteries, examined by immunohistochemistry. Osteogenic differentiation of primary human vascular smooth muscle cells increased DRP1 expression. DRP1 inhibition in human smooth muscle cells undergoing osteogenic differentiation attenuated matrix mineralization, cytoskeletal rearrangement, mitochondrial dysfunction, and reduced type 1 collagen secretion and alkaline phosphatase activity. DRP1 protein was observed in calcified human aortic valves, and DRP1 RNA interference reduced primary human valve interstitial cell calcification. Mice heterozygous for Drp1 deletion did not exhibit altered vascular pathology in a proprotein convertase subtilisin/kexin type 9 gain-of-function atherosclerosis model. However, when mineralization was induced via oxidative stress, DRP1 inhibition attenuated mouse and human smooth muscle cell calcification. Femur bone density was unchanged in mice heterozygous for Drp1 deletion, and DRP1 inhibition attenuated oxidative stress-mediated dysfunction in human bone osteoblasts. CONCLUSIONS: We demonstrate a new function of DRP1 in regulating collagen secretion and cardiovascular calcification, a novel area of exploration for the potential development of new therapies to modify cellular fibrocalcific response in cardiovascular diseases. Our data also support a role of mitochondrial dynamics in regulating oxidative stress-mediated arterial calcium accrual and bone loss.


Asunto(s)
GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/biosíntesis , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/biosíntesis , Miocitos del Músculo Liso/metabolismo , Estrés Oxidativo/fisiología , Calcificación Vascular/metabolismo , Calcificación Vascular/prevención & control , Animales , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/prevención & control , Células Cultivadas , Colágeno/metabolismo , Dinaminas , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Estrés Oxidativo/efectos de los fármacos , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Calcificación Vascular/patología
11.
Arterioscler Thromb Vasc Biol ; 38(1): 19-25, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29191923

RESUMEN

Cardiovascular disease is a leading cause of morbidity and mortality in the Western world. Studies of sortilin's influence on cardiovascular and metabolic diseases goes far beyond the genome-wide association studies that have revealed an association between cardiovascular diseases and the 1p13 locus that encodes sortilin. Emerging evidence suggests a significant role of sortilin in the pathogenesis of vascular and metabolic diseases; this includes type II diabetes mellitus via regulation of insulin resistance, atherosclerosis through arterial wall inflammation and calcification, and dysregulated lipoprotein metabolism. Sortilin is also known for its functional role in neurological disorders. It serves as a key receptor for cytokines, lipids, and enzymes and participates in pathological cargo loading to and trafficking of extracellular vesicles. This article provides a comprehensive review of sortilin's contributions to cardiovascular and metabolic diseases but focuses particularly on atherosclerosis. We summarize recent clinical findings that suggest that sortilin may be a cardiovascular risk biomarker and also discuss sortilin as a potential drug target.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Metabólicas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/genética , Diseño de Fármacos , Metabolismo Energético , Regulación de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/genética , Terapia Molecular Dirigida , Transducción de Señal
12.
Respir Res ; 19(1): 183, 2018 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-30236113

RESUMEN

BACKGROUND: Fibroblast growth factor 23 (FGF23) regulates phosphate metabolism by increasing renal phosphate excretion and decreasing 1.25-dihydroxyvitamin D synthesis. Reports about hypophosphatemia in patients with chronic obstructive pulmonary disease (COPD) suggest altered phosphate metabolism. Therefore, we hypothesized that disturbances in phosphate-regulatory hormones such as FGF23 and parathyroid hormone (PTH) are present in COPD patients. METHODS: We investigated 40 COPD patients (63.5 ± 9.9 years, 27 male), each matched with two age- and sex-matched controls without any primary lung disease. COPD patients underwent lung function testing in advance. All patients had a glomerular filtration rate (GFR) > 60 mL/min/1.73m2. We measured concentrations of intact FGF23 (iFGF23) and c-terminal FGF23 (c-term FGF23), phosphate, parathyroid hormone (PTH) and C-reactive protein (CRP) levels in COPD patients and controls. RESULTS: Phosphate (1.0 ± 02 vs. 1.1 ± 0.2 mmol/L; p = 0.027), PTH (54.2 ± 29.4 vs. 68.7 ± 31.8 pg/mL; p = 0.002) and iFGF23 (46.3 ± 29.0 vs. 57.5 ± 33.5 pg/mL; p = 0.026 ) levels were significantly lower in COPD patients compared with controls. There was a significant negative correlation between c-term FGF23 and total lung capacity (r = - 0.4; p = 0.01), and between c-term FGF23 and CRP in COPD patients (r = 0.48; p = 0.002). iFGF23 and c-term FGF23 were positively correlated with phosphate and PTH in the control group. CONCLUSION: We confirmed lower average serum phosphate levels in COPD patients compared with controls. However, our data do not suggest a causative role for FGF23 or PTH in COPD because levels of both phosphate-lowering hormones appear to be adaptively decreased as well. Therefore, further investigations are needed to identify the pathogenesis of low phosphate levels in patients with COPD and the relationship between phosphate-regulatory hormones and disease progression.


Asunto(s)
Factores de Crecimiento de Fibroblastos/sangre , Hormona Paratiroidea/sangre , Fosfatos/sangre , Enfermedad Pulmonar Obstructiva Crónica/sangre , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Anciano , Biomarcadores/sangre , Estudios de Casos y Controles , Estudios de Cohortes , Estudios Transversales , Femenino , Factor-23 de Crecimiento de Fibroblastos , Humanos , Masculino , Persona de Mediana Edad
13.
Arterioscler Thromb Vasc Biol ; 37(5): 1005-1011, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28279970

RESUMEN

OBJECTIVE: Genome-wide association studies and preclinical studies demonstrated a role of sortilin in lipid metabolism, inflammation, and vascular calcification-all cardiovascular risk factors. We evaluated the association of serum sortilin levels with the risk of major adverse cerebrovascular and cardiovascular events (MACCE) and the severity of abdominal aortic calcification (AAC). APPROACH AND RESULTS: A cohort of community-dwelling men aged ≥50 years (n=830) was assessed. At baseline, sortilin levels were measured by ELISA, and AAC was assessed on lateral spine scans obtained by dual-energy X-ray absorptiometry. Men aged ≥60 years (n=745) were followed up prospectively for the incidence of MACCE. During the median follow-up of 7.9 years, 76 MACCE occurred. The unadjusted incidence of MACCE across increasing sortilin quartiles was 8.0, 7.4, 19.8, and 20.3 per 1000 person-years. In multivariate-adjusted analysis, sortilin associated with increased risk of MACCE (hazard ratio, 1.70 per SD; 95% confidence interval, 1.30-2.20; P<0.001). The third and fourth quartiles associated with 3.42-fold (95% confidence interval, 1.61-7.25; P<0.005) and 3.82-fold (95% confidence interval, 1.77-8.26; P<0.001) higher risk of MACCE compared with the first quartile. High sortilin also predicted MACCE independent of traditional Framingham risk factors. Higher sortilin associated with higher odds of severe AAC (score>5) after adjustment for confounders (odds ratio, 1.43 per SD; 95% confidence interval, 1.10-1.85; P<0.01). The highest sortilin quartile associated with 2-fold higher odds of severe AAC (versus 3 lower quartiles combined). After adjustment for low-density lipoprotein cholesterol, the odds of severe AAC remained significant. CONCLUSIONS: In older men, higher serum sortilin levels associated with higher MACCE risk and severe AAC independently of relevant confounders, including C-reactive protein and low-density lipoprotein cholesterol. This finding, however, needs to be validated in other cohorts.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/sangre , Aorta Abdominal , Enfermedades de la Aorta/sangre , Trastornos Cerebrovasculares/sangre , Cardiopatías/sangre , Calcificación Vascular/sangre , Absorciometría de Fotón , Anciano , Anciano de 80 o más Años , Aorta Abdominal/diagnóstico por imagen , Enfermedades de la Aorta/diagnóstico por imagen , Enfermedades de la Aorta/epidemiología , Biomarcadores/sangre , Proteína C-Reactiva/análisis , Trastornos Cerebrovasculares/diagnóstico , Trastornos Cerebrovasculares/epidemiología , Distribución de Chi-Cuadrado , LDL-Colesterol/sangre , Ensayo de Inmunoadsorción Enzimática , Francia/epidemiología , Cardiopatías/diagnóstico , Cardiopatías/epidemiología , Humanos , Incidencia , Estimación de Kaplan-Meier , Modelos Lineales , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Oportunidad Relativa , Pronóstico , Estudios Prospectivos , Medición de Riesgo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Factores Sexuales , Factores de Tiempo , Regulación hacia Arriba , Calcificación Vascular/diagnóstico por imagen , Calcificación Vascular/epidemiología
14.
Semin Cell Dev Biol ; 46: 68-77, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26358815

RESUMEN

The presence of cardiovascular calcification significantly predicts patients' morbidity and mortality. Calcific mineral deposition within the soft cardiovascular tissues disrupts the normal biomechanical function of these tissues, leading to complications such as heart failure, myocardial infarction, and stroke. The realization that calcification results from active cellular processes offers hope that therapeutic intervention may prevent or reverse the disease. To this point, however, no clinically viable therapies have emerged. This may be due to the lack of certainty that remains in the mechanisms by which mineral is deposited in cardiovascular tissues. Gaining new insight into this process requires a multidisciplinary approach. The pathological changes in cell phenotype that lead to the physicochemical deposition of mineral and the resultant effects on tissue biomechanics must all be considered when designing strategies to treat cardiovascular calcification. In this review, we overview the current cardiovascular calcification paradigm and discuss emerging techniques that are providing new insight into the mechanisms of ectopic calcification.


Asunto(s)
Calcinosis/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Placa Aterosclerótica/metabolismo , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Válvula Aórtica/fisiopatología , Enfermedades Cardiovasculares/patología , Colágeno/metabolismo , Enfermedad de la Arteria Coronaria/patología , Humanos , Inflamación/metabolismo , Modelos Biológicos , Placa Aterosclerótica/patología
15.
Nat Mater ; 15(3): 335-43, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26752654

RESUMEN

Clinical evidence links arterial calcification and cardiovascular risk. Finite-element modelling of the stress distribution within atherosclerotic plaques has suggested that subcellular microcalcifications in the fibrous cap may promote material failure of the plaque, but that large calcifications can stabilize it. Yet the physicochemical mechanisms underlying such mineral formation and growth in atheromata remain unknown. Here, by using three-dimensional collagen hydrogels that mimic structural features of the atherosclerotic fibrous cap, and high-resolution microscopic and spectroscopic analyses of both the hydrogels and of calcified human plaques, we demonstrate that calcific mineral formation and maturation results from a series of events involving the aggregation of calcifying extracellular vesicles, and the formation of microcalcifications and ultimately large calcification areas. We also show that calcification morphology and the plaque's collagen content-two determinants of atherosclerotic plaque stability-are interlinked.


Asunto(s)
Aterosclerosis/metabolismo , Vesículas Extracelulares/fisiología , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Calcio/metabolismo , Arterias Carótidas/patología , Colágeno/metabolismo , Enfermedad Coronaria/metabolismo , Matriz Extracelular , Humanos , Ratones , Ratones Noqueados
16.
J Mol Cell Cardiol ; 94: 13-20, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26996755

RESUMEN

PURPOSE: Calcific aortic valve disease (CAVD) is the most prevalent valve disease in the Western world. Recent difficulty in translating experimental results on statins to beneficial clinical effects warrants the need for understanding the role of valvular interstitial cells (VICs) in CAVD. In two-dimensional culture conditions, VICs undergo spontaneous activation similar to pathological differentiation, which intrinsically limits the use of in vitro models to study CAVD. Here, we hypothesized that a three-dimensional (3D) culture system based on naturally derived extracellular matrix polymers, mimicking the microenvironment of native valve tissue, could serve as a physiologically relevant platform to study the osteogenic differentiation of VICs. PRINCIPAL RESULTS: Aortic VICs loaded into 3D hydrogel constructs maintained a quiescent phenotype, similar to healthy human valves. In contrast, osteogenic environment induced an initial myofibroblast differentiation (hallmarked by increased alpha smooth muscle actin [α-SMA] expression), followed by an osteoblastic differentiation, characterized by elevated Runx2 expression, and subsequent calcific nodule formation recapitulating CAVD conditions. Silencing of α-SMA under osteogenic conditions diminished VIC osteoblast-like differentiation and calcification, indicating that a VIC myofibroblast-like phenotype may precede osteogenic differentiation in CAVD. MAJOR CONCLUSIONS: Using a 3D hydrogel model, we simulated events that occur during early CAVD in vivo and provided a platform to investigate mechanisms of CAVD. Differentiation of valvular interstitial cells to myofibroblasts was a key mechanistic step in the process of early mineralization. This novel approach can provide important insight into valve pathobiology and serve as a promising tool for drug screening.


Asunto(s)
Estenosis de la Válvula Aórtica/etiología , Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Calcinosis/etiología , Calcinosis/metabolismo , Actinas/genética , Animales , Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/patología , Apoptosis , Biomarcadores , Calcinosis/genética , Calcinosis/patología , Técnicas de Cultivo de Célula , Ciclo Celular , Diferenciación Celular , Supervivencia Celular , Técnica del Anticuerpo Fluorescente , Silenciador del Gen , Humanos , Hidrogeles , Técnicas In Vitro , Miofibroblastos/citología , Miofibroblastos/metabolismo , Fenotipo , Porcinos
17.
Circ Res ; 112(7): 1073-84, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23538277

RESUMEN

Cardiovascular calcification is a prominent feature of chronic inflammatory disorders-such as chronic kidney disease, type 2 diabetes mellitus, and atherosclerosis-that associate with significant morbidity and mortality. The concept that similar pathways control both bone remodeling and vascular calcification is widely accepted, but the precise mechanisms of calcification remain largely unknown. The central role of microRNAs (miRNA) as fine-tune regulators in the cardiovascular system and bone biology has gained acceptance and has raised the possibility for novel therapeutic targets. Additionally, circulating miRNAs have been proposed as biomarkers for a wide range of cardiovascular diseases, but knowledge of miRNA biology in cardiovascular calcification is very limited. This review focuses on the role of miRNAs in cardiovascular disease, with emphasis on osteogenic processes. Herein, we discuss the current understanding of miRNAs in cardiovascular calcification. Furthermore, we identify a set of miRNAs common to diseases associated with cardiovascular calcification (chronic kidney disease, type 2 diabetes mellitus, and atherosclerosis), and we hypothesize that these miRNAs may provide a molecular signature for calcification. Finally, we discuss this novel hypothesis with emphasis on known biological and pathological osteogenic processes (eg, osteogenic differentiation, release of calcifying matrix vesicles). The aim of this review is to provide an organized discussion of the known links between miRNA and calcification that provide emerging concepts for future studies on miRNA biology in cardiovascular calcification, which will be critical for developing new therapeutic strategies.


Asunto(s)
Calcinosis/genética , Calcinosis/terapia , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/terapia , Terapia Genética/métodos , MicroARNs/fisiología , Animales , Calcinosis/patología , Enfermedades Cardiovasculares/patología , Sistemas de Liberación de Medicamentos , Humanos
18.
Circ Res ; 113(1): 72-7, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23616621

RESUMEN

RATIONALE: We previously showed that early calcification of atherosclerotic plaques associates with macrophage accumulation. Chronic renal disease and mineral imbalance accelerate calcification and the subsequent release of matrix vesicles (MVs), precursors of microcalcification. OBJECTIVE: We tested the hypothesis that macrophage-derived MVs contribute directly to microcalcification. METHODS AND RESULTS: Macrophages associated with regions of calcified vesicular structures in human carotid plaques (n=136 patients). In vitro, macrophages released MVs with high calcification and aggregation potential. MVs expressed exosomal markers (CD9 and TSG101) and contained S100A9 and annexin V. Silencing S100A9 in vitro and genetic deficiency in S100A9-/- mice reduced MV calcification, whereas stimulation with S100A9 increased calcification potential. Externalization of phosphatidylserine after Ca/P stimulation and interaction of S100A9 and annexin V indicated that a phosphatidylserine-annexin V-S100A9 membrane complex facilitates hydroxyapatite nucleation within the macrophage-derived MV membrane. CONCLUSIONS: Our results support the novel concept that macrophages release calcifying MVs enriched in S100A9 and annexin V, which contribute to accelerated microcalcification in chronic renal disease.


Asunto(s)
Anexina A5/metabolismo , Calcinosis/metabolismo , Calgranulina B/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Vesículas Citoplasmáticas/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica/metabolismo , Animales , Apolipoproteínas E/deficiencia , Calcinosis/patología , Calcio/farmacología , Calgranulina B/genética , Enfermedades de las Arterias Carótidas/patología , Línea Celular , Vesículas Citoplasmáticas/ultraestructura , Durapatita/metabolismo , Humanos , Macrófagos/ultraestructura , Macrófagos Peritoneales/fisiología , Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilserinas/metabolismo , Fósforo/farmacología , Placa Aterosclerótica/patología , Interferencia de ARN , ARN Interferente Pequeño/farmacología
19.
Arterioscler Thromb Vasc Biol ; 34(3): 626-34, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24357058

RESUMEN

OBJECTIVE: Clinical evidence has linked vascular calcification in advanced atherosclerotic plaques with overt cardiovascular disease and mortality. Bone resorbing monocyte-derived osteoclast-like cells are sparse in these plaques, indicating that their differentiation capability could be suppressed. Here, we seek to characterize the process of osteoclastogenesis by identifying novel regulators and pathways, with the aim of exploring possible strategies to reduce calcification. APPROACH AND RESULTS: We used a quantitative mass spectrometry strategy, tandem mass tagging, to quantify changes in the proteome of osteoclast-like cells differentiated from RAW264.7 cells in response to, receptor activator of nuclear factor κ-B ligand induction, a common in vitro model for osteogenesis. More than 4000 proteins were quantified, of which 138 were identified as novel osteoclast-related proteins. We selected 5 proteins for subsequent analysis (cystathionine γ-lyase [Cth/CSE], EGF-like repeat and discoidin I-like domain-containing protein 3, integrin α FG-GAP repeat containing 3, adseverin, and serpinb6b) and show that gene expression levels are also increased. Further analysis of the CSE transcript profile reveals an early onset of an mRNA increase. Silencing of CSE by siRNA and dl-propargylglycine, a CSE inhibitor, attenuated receptor activator of nuclear factor κ-B ligand-induced tartrate-resistant acid phosphatase type 5 activity and pit formation, suggesting that CSE is a potent inducer of calcium resorption. Moreover, knockdown of CSE suppressed expression of osteoclast differentiation markers. CONCLUSIONS: Our large-scale proteomics study identified novel candidate regulators or markers for osteoclastogenesis and demonstrated that CSE may act in early stages of osteoclastogenesis.


Asunto(s)
Cistationina gamma-Liasa/fisiología , Osteoclastos/enzimología , Alquinos/farmacología , Animales , Aorta Torácica/metabolismo , Apolipoproteínas E/deficiencia , Biomarcadores , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular , Línea Celular Tumoral , Cistationina gamma-Liasa/antagonistas & inhibidores , Cistationina gamma-Liasa/genética , Grasas de la Dieta/toxicidad , Perfilación de la Expresión Génica , Glicina/análogos & derivados , Glicina/farmacología , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Proteómica , Ligando RANK/farmacología , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/farmacología , Espectrometría de Masa por Ionización de Electrospray , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA