Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Calcif Tissue Int ; 115(3): 315-327, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38951181

ABSTRACT

Vascular calcification affects the prognosis of patients with renal failure. Bisphosphonates are regarded as candidate anti-calcifying drugs because of their inhibitory effects on both calcium-phosphate aggregation and bone resorption. However, calcification in well-known rodent models is dependent upon bone resorption accompanied by excessive bone turnover, making it difficult to estimate accurately the anti-calcifying potential of drugs. Therefore, models with low bone resorption are required to extrapolate anti-calcifying effects to humans. Three bisphosphonates (etidronate, alendronate, and FYB-931) were characterised for their inhibitory effects on bone resorption in vivo and calcium-phosphate aggregation estimated by calciprotein particle formation in vitro. Then, their effects were examined using two models inducing ectopic calcification: the site where lead acetate was subcutaneously injected into mice and the transplanted, aorta obtained from a donor rat. The inhibitory effects of bisphosphonates on bone resorption and calcium-phosphate aggregation were alendronate > FYB-931 > etidronate and FYB-931 > alendronate = etidronate, respectively. In the lead acetate-induced model, calcification was most potently suppressed by FYB-931, followed by alendronate and etidronate. In the aorta-transplanted model, only FYB-931 suppressed calcification at a high dose. In both the models, no correlation was observed between calcification and bone resorption marker, tartrate-resistant acid phosphatase (TRACP). Results from the lead acetate-induced model showed that inhibitory potency against calcium-phosphate aggregation contributed to calcification inhibition. The two calcification models, especially the lead acetate-induced model, may be ideal for the extrapolation of calcifying response to humans because of calcium-phosphate aggregation rather than bone resorption as its mechanism.


Subject(s)
Bone Resorption , Disease Models, Animal , Organometallic Compounds , Animals , Mice , Humans , Organometallic Compounds/pharmacology , Rats , Diphosphonates/pharmacology , Male , Bone Density Conservation Agents/pharmacology , Alendronate/pharmacology , Calcinosis/chemically induced , Mice, Inbred C57BL , Vascular Calcification/chemically induced
2.
Cardiovasc Res ; 120(7): 699-707, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38636937

ABSTRACT

Despite the air quality has been generally improved in recent years, ambient fine particulate matter (PM2.5), a major contributor to air pollution, remains one of the major threats to public health. Vascular calcification is a systematic pathology associated with an increased risk of cardiovascular disease. Although the epidemiological evidence has uncovered the association between PM2.5 exposure and vascular calcification, little is known about the underlying mechanisms. The adverse outcome pathway (AOP) concept offers a comprehensive interpretation of all of the findings obtained by toxicological and epidemiological studies. In this review, reactive oxygen species generation was identified as the molecular initiating event (MIE), which targeted subsequent key events (KEs) such as oxidative stress, inflammation, endoplasmic reticulum stress, and autophagy, from the cellular to the tissue/organ level. These KEs eventually led to the adverse outcome, namely increased incidence of vascular calcification and atherosclerosis morbidity. To the best of our knowledge, this is the first AOP framework devoted to PM2.5-associated vascular calcification, which benefits future investigations by identifying current limitations and latent biomarkers.


Subject(s)
Air Pollutants , Oxidative Stress , Particulate Matter , Vascular Calcification , Particulate Matter/adverse effects , Humans , Vascular Calcification/metabolism , Vascular Calcification/epidemiology , Vascular Calcification/pathology , Vascular Calcification/chemically induced , Animals , Air Pollutants/adverse effects , Oxidative Stress/drug effects , Risk Factors , Risk Assessment , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/metabolism , Reactive Oxygen Species/metabolism , Environmental Exposure/adverse effects , Air Pollution/adverse effects , Autophagy/drug effects , Inflammation Mediators/metabolism , Particle Size , Prognosis , Endoplasmic Reticulum Stress/drug effects , Signal Transduction
3.
Kidney Blood Press Res ; 49(1): 137-143, 2024.
Article in English | MEDLINE | ID: mdl-38266504

ABSTRACT

INTRODUCTION: The process of vascular calcification has severe clinical consequences in a number of diseases, including diabetes, atherosclerosis, and end-stage renal disease. In the present study, we investigated the effect of policosanol (Poli), genistein (Gen), and vitamin D (VitD) separately and in association to evaluate the possible synergistic action on inorganic phosphate (Pi)-induced calcification of vascular smooth muscle cells (VSMCs). METHODS: Primary human VSMCs were cultured with either growth medium or growth medium supplemented with calcium and phosphorus (calcification medium) in combination with Poli, Gen, and VitD. Alizarin Red staining, mineralization, and the protein expression of RUNX2 and superoxide dismutase-2 (SOD2) were investigated. RESULTS: All three substances tested were effective at reducing osteogenic differentiation of VSMCs in a dose-dependent manner. Poli+Gen, Poli+VitD, Gen+VitD treatment induced a greater inhibition of calcification and RUNX2 expression compared to single compounds treatments. Moreover, the association of Poli+Gen+VitD (Reduplaxin®) was more effective at inhibiting VSMCs mineralization and preventing the increase in RUNX2 expression induced by calcification medium but not modified SOD2 expression. CONCLUSIONS: The association of Pol, Gen, and VitD (Reduplaxin®) has an additive inhibitory effect on the calcification process of VSMCs induced in vitro by a pro-calcifying medium.


Subject(s)
Fatty Alcohols , Genistein , Muscle, Smooth, Vascular , Vascular Calcification , Vitamin D , Humans , Vitamin D/pharmacology , Fatty Alcohols/pharmacology , Cells, Cultured , Vascular Calcification/prevention & control , Vascular Calcification/chemically induced , Vascular Calcification/drug therapy , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Genistein/pharmacology , Genistein/therapeutic use , Superoxide Dismutase/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism
4.
Biomolecules ; 14(1)2023 Dec 24.
Article in English | MEDLINE | ID: mdl-38254629

ABSTRACT

Vascular calcification (VC) is an age-related complication characterised by calcium-phosphate deposition in the arterial wall driven by the osteogenic transformation of vascular smooth muscle cells (VSMCs). The JAK-STAT pathway is an emerging target in inflammation. Considering the relationship between VC and inflammation, we investigated the role of JAK-STAT signalling during VSMC calcification. Human aortic smooth muscle cells (HASMCs) were cultured in high-inorganic phosphate (Pi) medium for up to 7 days; calcium deposition was determined via Alizarin staining and colorimetric assay. Inflammatory factor secretion was evaluated via ELISA and JAK-STAT members' activation using Western blot or immunohistochemistry on HASMCs or calcified aortas of Vitamin D-treated C57BL6/J mice, respectively. The JAK-STAT pathway was blocked by JAK Inhibitor I and Von Kossa staining was used for calcium deposits in murine aortic rings. During Pi-induced calcification, HASMCs released IL-6, IL-8, and MCP-1 and activated JAK1-JAK3 proteins and STAT1. Phospho-STAT1 was detected in murine calcified aortas. Blocking of the JAK-STAT cascade reduced HASMC proliferation and pro-inflammatory factor expression and release while increasing calcium deposition and osteogenic transcription factor RUNX2 expression. Consistently, JAK-STAT pathway inhibition exacerbates mouse aortic ring calcification ex vivo. Intriguingly, our results suggest an alternative link between VSMC inflammation and VC.


Subject(s)
Muscle, Smooth, Vascular , Vascular Calcification , Humans , Animals , Mice , Calcium , Janus Kinases , STAT Transcription Factors , Signal Transduction , Vascular Calcification/chemically induced , Inflammation
5.
Angiología ; 68(2): 96-103, mar.-abr. 2016. ilus, tab
Article in Spanish | IBECS | ID: ibc-148294

ABSTRACT

OBJETIVO: Nanopartículas calcificantes (NP) se han detectado recientemente en muestras arteriales humanas y parecen estar involucradas en el proceso de calcificación. Este estudio fue diseñado para probar la hipótesis de que las NP de origen humano podrían agravar la respuesta a la lesión arterial endotelial e inducir la calcificación vascular. MÉTODOS: La arteria carótida derecha de 24 conejos neozelandeses fue lesionada con un balón de angioplastia. Los animales fueron perfundidos por vía intravenosa con solución salina (100 ml) durante el experimento y se dividieron en 3 grupos: grupo A, control; grupo B, expuesto a NP (2 ml) obtenidas a partir de válvulas aórticas calcificadas y el grupo C, expuesto a NP (2 ml) y tratado después de la operación con atorvastatina (2,5 mg/kg/24 h). A los 30 días, los animales fueron sacrificados y se extirparon las 2 arterias carótidas, que fueron examinadas histológicamente. Análisis bioquímicos de sangre fueron realizados durante el estudio. RESULTADOS: El área de hiperplasia intimal fue significativamente mayor en la arteria carótida derecha lesionada en comparación con la arteria carótida izquierda no operada, en todos los grupos. No hubo variación significativa en la zona medial entre los animales. Morfométricamente, la relación de íntima/media (IMR) fue significativamente mayor en las carótidas dañadas en comparación con los controles. Un aumento significativo de IMR se encontró en el grupo B (1,81 ± 0,41) en comparación con el grupo A (0,38 ± 0,59; p = 0,004) o el grupo C (0,89 ± 0,79; p = 0,035). Las diferencias entre los grupos C y A no fueron significativas (p = 0,064). Se observaron calcificaciones en 6 animales, todos los cuales habían sido expuestos a NP (4 en el grupo B, 2 en el grupo C, p = 0,027). Los niveles plasmáticos de colesterol y triglicéridos se mantuvieron estables. CONCLUSIONES: Este estudio confirma la capacidad de las NP de origen humano de acelerar la hiperplasia y estimular la calcificación de zonas arteriales endoteliales previamente dañadas. Su administración sistémica resultó inofensiva en las arterias sanas. La atorvastatina demostró la capacidad de ralentizar este proceso


OBJECTIVE: Calcifying nanoparticles (NP) have been detected recently in calcified human arterial specimens, and are involved in the process of calcification. This study was designed to test the hypothesis that human-derived NP could worsen the response to arterial endothelial injury and induce vascular calcification. METHODS: The right carotid artery of 24 New Zealand rabbits was injured with an angioplasty balloon. Animals were perfused intravenously with saline (100 mL) during the experiment and divided into 3 groups: group A, control; group B, exposed to NP (2 mL) obtained from calcified aortic valves; and group C, exposed to NP (2 mL) and treated post-operatively with atorvastatin (2.5 mg/kg/24 h). At 30 days, both carotid arteries were removed and examined histologically. Blood measurements were monitored during the study. RESULTS: The intimal hyperplasia area was significantly larger in the injured right carotid artery compared with the left un-operated carotid artery in all groups. There was no significant variation in medial area between groups. Morphometrically, the intima/media ratio (IMR) was significantly higher in damaged carotids compared with controls. A significant increase in the IMR was found in group B (1.81 ± 0.41) compared with group A (0.38 ± 0.59; P=.004) or group C (0.89 ± 0.79; P=.035). Differences between groups C and A were not significant (P=.064). Calcifications were observed in 6 animals, all of which had been exposed to NP (4 in group B, and 2 in group C, P=.027). Plasma levels of cholesterol and triglycerides remained stable. CONCLUSIONS: his study confirms the ability of systemic inoculation of human-derived NP to accelerate hyperplasia and stimulate calcification in localised areas of arteries previously submitted to endothelial damage, while it was harmless in healthy arteries. Atorvastatin was demonstrated to slow down this process


Subject(s)
Animals , Male , Female , Rabbits , Calcifying Nanoparticles/analysis , Calcifying Nanoparticles , Hyperplasia/diagnosis , Hyperplasia/ethnology , Vascular Calcification/chemically induced , Vascular Calcification/complications , Vascular Calcification/veterinary , Carotid Artery Injuries/drug therapy , Carotid Artery Injuries/veterinary , Atherosclerosis/drug therapy , Atherosclerosis/veterinary
6.
Braz. j. med. biol. res ; 47(2): 119-127, 2/2014. graf
Article in English | LILACS | ID: lil-699771

ABSTRACT

Vascular calcification decreases compliance and increases morbidity. Mechanisms of this process are unclear. The role of oxidative stress and effects of antioxidants have been poorly explored. We investigated effects of the antioxidants lipoic acid (LA) and tempol in a model of atherosclerosis associated with elastocalcinosis. Male New Zealand white rabbits (2.5-3.0 kg) were fed regular chow (controls) or a 0.5% cholesterol (chol) diet+104 IU/day vitamin D2 (vitD) for 12 weeks, and assigned to treatment with water (vehicle, n=20), 0.12 mmol·kg-1·day-1 LA (n=11) or 0.1 mmol·kg-1·day-1 tempol (n=15). Chol+vitD-fed rabbits developed atherosclerotic plaques associated with expansive remodeling, elastic fiber disruption, medial calcification, and increased aortic stiffness. Histologically, LA prevented medial calcification by ∼60% and aortic stiffening by ∼60%. LA also preserved responsiveness to constrictor agents, while intima-media thickening was increased. In contrast to LA, tempol was associated with increased plaque collagen content, medial calcification and aortic stiffness, and produced differential changes in vasoactive responses in the chol+vitD group. Both LA and tempol prevented superoxide signals with chol+vitD. However, only LA prevented hydrogen peroxide-related signals with chol+vitD, while tempol enhanced them. These data suggest that LA, opposite to tempol, can minimize calcification and compliance loss in elastocalcionosis by inhibition of hydrogen peroxide generation.


Subject(s)
Animals , Male , Rabbits , Arteriosclerosis/prevention & control , Cyclic N-Oxides/administration & dosage , Thioctic Acid/administration & dosage , Vascular Calcification/prevention & control , Aorta, Thoracic , Arteriosclerosis/chemically induced , Arteriosclerosis/metabolism , Compliance/drug effects , Compliance/physiology , Disease Models, Animal , Spin Labels , Vascular Resistance , Vascular Calcification/chemically induced , Vasoconstriction/drug effects , Vasoconstriction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL