Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
FASEB J ; 38(7): e23592, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38581243

RESUMEN

Vascular calcification is an actively regulated biological process resembling bone formation, and osteogenic differentiation of vascular smooth muscle cells (VSMCs) plays a crucial role in this process. 1-Palmitoyl-2-(5'-oxo-valeroyl)-sn-glycero-3-phosphocholine (POVPC), an oxidized phospholipid, is found in atherosclerotic plaques and has been shown to induce oxidative stress. However, the effects of POVPC on osteogenic differentiation and calcification of VSMCs have yet to be studied. In the present study, we investigated the role of POVPC in vascular calcification using in vitro and ex vivo models. POVPC increased mineralization of VSMCs and arterial rings, as shown by alizarin red staining. In addition, POVPC treatment increased expression of osteogenic markers Runx2 and BMP2, indicating that POVPC promotes osteogenic transition of VSMCs. Moreover, POVPC increased oxidative stress and impaired mitochondria function of VSMCs, as shown by increased ROS levels, impairment of mitochondrial membrane potential, and decreased ATP levels. Notably, ferroptosis triggered by POVPC was confirmed by increased levels of intracellular ROS, lipid ROS, and MDA, which were decreased by ferrostatin-1, a ferroptosis inhibitor. Furthermore, ferrostatin-1 attenuated POVPC-induced calcification of VSMCs. Taken together, our study for the first time demonstrates that POVPC promotes vascular calcification via activation of VSMC ferroptosis. Reducing the levels of POVPC or inhibiting ferroptosis might provide a novel strategy to treat vascular calcification.


Asunto(s)
Ciclohexilaminas , Ferroptosis , Fenilendiaminas , Calcificación Vascular , Humanos , Músculo Liso Vascular/metabolismo , Fosfolípidos/metabolismo , Fosforilcolina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Osteogénesis , Calcificación Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Células Cultivadas
2.
J Pathol ; 258(3): 213-226, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35894849

RESUMEN

Vascular calcification is an actively regulated process resembling bone formation and contributes to the cardiovascular morbidity and mortality of chronic kidney disease (CKD). However, an effective therapy for vascular calcification is still lacking. The ketone body ß-hydroxybutyrate (BHB) has been demonstrated to have health-promoting effects including anti-inflammation and cardiovascular protective effects. However, whether BHB protects against vascular calcification in CKD remains unclear. In this study, Alizarin Red staining and calcium content assay showed that BHB reduced calcification of vascular smooth muscle cells (VSMCs) and arterial rings. Of note, compared with CKD patients without thoracic calcification, serum BHB levels were lower in CKD patients with thoracic calcification. Supplementation with 1,3-butanediol (1,3-B), the precursor of BHB, attenuated aortic calcification in CKD rats and VitD3-overloaded mice. Furthermore, RNA-seq analysis revealed that BHB downregulated HDAC9, which was further confirmed by RT-qPCR and western blot analysis. Both pharmacological inhibition and knockdown of HDAC9 attenuated calcification of human VSMCs, while overexpression of HDAC9 exacerbated calcification of VSMCs and aortic rings, indicating that HDAC9 promotes vascular calcification under CKD conditions. Of note, BHB treatment antagonized HDAC9-induced vascular calcification. In addition, HDAC9 overexpression activated the NF-κB signaling pathway and inhibition of NF-κB attenuated HDAC9-induced VSMC calcification, suggesting that HDAC9 promotes vascular calcification via activation of NF-κB. In conclusion, our study demonstrates that BHB supplementation inhibits vascular calcification in CKD via modulation of the HDAC9-dependent NF-κB signaling pathway. Moreover, we unveil a crucial mechanistic role of HDAC9 in vascular calcification under CKD conditions; thus, nutritional intervention or pharmacological approaches to enhance BHB levels could act as promising therapeutic strategies to target HDAC9 for the treatment of vascular calcification in CKD. © 2022 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Insuficiencia Renal Crónica , Calcificación Vascular , Ácido 3-Hidroxibutírico/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Regulación hacia Abajo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Cetonas/metabolismo , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/patología , FN-kappa B/metabolismo , Ratas , Insuficiencia Renal Crónica/patología , Proteínas Represoras/metabolismo , Calcificación Vascular/genética , Calcificación Vascular/prevención & control
3.
J Mol Cell Cardiol ; 173: 154-168, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36367517

RESUMEN

Vascular calcification is an important risk factor for cardiovascular events, accompanied by DNA damage during the process. The sirtuin 6 (SIRT6) has been reported to alleviate atherosclerosis, which is related to the reduction of DNA damage. However, whether smooth muscle cell SIRT6 mediates vascular calcification involving DNA damage remains unclear. Western blot and immunofluorescence revealed that SIRT6 expression was decreased in human vascular smooth muscle cells (HVSMCs), human and mouse arteries during vascular calcification. Alizarin red staining and calcium content assay showed that knockdown or deletion of SIRT6 significantly promoted HVSMC calcification induced by high phosphorus and calcium, accompanied by upregulation of osteogenic differentiation markers including Runx2 and BMP2. By contrast, adenovirus-mediated SIRT6 overexpression attenuated osteogenic differentiation and calcification of HVSMCs. Moreover, ex vivo study revealed that SIRT6 overexpression inhibited calcification of mouse and human arterial rings. Of note, smooth muscle cell-specific knockout of SIRT6 markedly aggravated Vitamin D3-induced aortic calcification in mice. Mechanistically, overexpression of SIRT6 reduced DNA damage and upregulated p-ATM during HVSMCs calcification, whereas knockdown of SIRT6 showed the opposite effects. Knockdown of ATM in HVSMCs abrogated the inhibitory effect of SIRT6 overexpression on calcification and DNA damage. This study for the first time demonstrates that vascular smooth muscle cell-specific deletion of SIRT6 facilitates vascular calcification via suppression of DNA damage repair. Therefore, modulation of SIRT6 and DNA damage repair may represent a therapeutic strategy for vascular calcification.


Asunto(s)
Sirtuinas , Calcificación Vascular , Humanos , Calcio/metabolismo , Daño del ADN , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Osteogénesis/genética , Sirtuinas/genética , Sirtuinas/metabolismo , Calcificación Vascular/genética , Reparación del ADN
4.
Kidney Int ; 102(6): 1259-1275, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36063875

RESUMEN

Vascular calcification is a common pathologic condition in patients with chronic kidney disease (CKD). Cell death such as apoptosis plays a critical role in vascular calcification. Ferroptosis is a type of iron-catalyzed and regulated cell death resulting from excessive iron-dependent reactive oxygen species and lipid peroxidation. However, it is unclear whether ferroptosis of vascular smooth muscle cells (VSMCs) regulates vascular calcification in CKD. Our results showed that high calcium and phosphate concentrations induced ferroptosis in rat VSMCs in vitro. Inhibition of ferroptosis by ferrostatin-1 dose-dependently reduced mineral deposition in rat VSMCs under pro-osteogenic conditions, as indicated by alizarin red staining and quantification of calcium content. In addition, gene expression analysis revealed that ferrostatin-1 inhibited osteogenic differentiation of rat VSMCs. Similarly, ferrostatin-1 remarkably attenuated calcification of rat and human arterial rings ex vivo and aortic calcification in vitamin D3-overloaded mice in vivo. Moreover, inhibition of ferroptosis by either ferrostatin-1 or deferoxamine attenuated aortic calcification in rats with CKD. Mechanistically, high calcium and phosphate downregulated expression of SLC7A11 (a cystine-glutamate antiporter), and reduced glutathione (GSH) content in VSMCs. Additionally, GSH depletion induced by erastin (a small molecule initiating ferroptotic cell death) significantly promoted calcification of VSMCs under pro-osteogenic conditions, whereas GSH supplement by N-acetylcysteine reduced calcification of VSMCs. Consistently, knockdown of SLC7A11 by siRNA markedly promoted VSMC calcification. Furthermore, high calcium and phosphate downregulated glutathione peroxidase 4 (GPX4) expression, and reduced glutathione peroxidase activity. Inhibition of GPX4 by RSL3 promoted VSMC calcification. Thus, repression of the SLC7A11/GSH/GPX4 axis triggers ferroptosis of VSMCs to promote vascular calcification under CKD conditions, providing a novel targeting strategy for vascular calcification.


Asunto(s)
Ferroptosis , Insuficiencia Renal Crónica , Calcificación Vascular , Humanos , Ratas , Ratones , Animales , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Músculo Liso Vascular , Osteogénesis , Calcio/metabolismo , Antiportadores/metabolismo , Miocitos del Músculo Liso/metabolismo , Calcificación Vascular/genética , Calcificación Vascular/prevención & control , Hierro/metabolismo , Glutatión/metabolismo , Insuficiencia Renal Crónica/patología , Fosfatos/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo
5.
Int Heart J ; 63(5): 928-938, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36184552

RESUMEN

The role of endothelial injury and inflammation in atherosclerosis has been well established. miRNAs have been found to be key regulators in the development of atherosclerosis. Here we investigated whether miR-32-5p and its predicted target gene axin interactor, dorsalization associated (AIDA) are involved in endothelial injury and inflammation. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (oxLDL) to induce endothelial injury and inflammation. AIDA was predicted to be a target gene of miR-32-5p using TargetScan software. Cell viability, migration, and angiogenesis were evaluated using Cell Counting Kit-8, wound-healing, and tube formation assays, respectively. The expression of inflammatory factors was detected using quantitative PCR, enzyme-linked immunosorbent assay, and western blot. We found that miR-32-5p expression was significantly decreased, whereas AIDA expression was significantly increased in oxLDL-treated HUVECs and the increased AIDA expression was reversed by the up-regulation of miR-32-5p. Moreover, both miR-32-5p mimic and knockdown of AIDA enhanced cell viability, promoted cell migration and angiogenesis and suppressed the expression of inflammatory factors including IL-1ß, IL-6, TNF-α, ICAM-1, and VCAM-1 in oxLDL-induced HUVECs. Furthermore, miR-32-5p was verified to directly target AIDA using dual-luciferase reporter assay. Overall, these findings suggest that miR-32-5p/AIDA signal plays an important role in oxLDL-induced endothelial injury and inflammation. This study provides new insights into novel molecular mechanisms of endothelial dysfunction and atherosclerosis.


Asunto(s)
Aterosclerosis , MicroARNs , Apoptosis , Aterosclerosis/genética , Aterosclerosis/metabolismo , Proteína Axina/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/metabolismo , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Transferencia de Fosfolípidos , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular
6.
Arterioscler Thromb Vasc Biol ; 40(3): 751-765, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31941382

RESUMEN

OBJECTIVES: Vascular calcification is highly prevalent in patients with chronic kidney disease. Increased plasma trimethylamine N-oxide (TMAO), a gut microbiota-dependent product, concentrations are found in patients undergoing hemodialysis. However, a clear mechanistic link between TMAO and vascular calcification is not yet established. In this study, we investigate whether TMAO participates in the progression of vascular calcification using in vitro, ex vivo, and in vivo models. Approach and Results: Alizarin red staining revealed that TMAO promoted calcium/phosphate-induced calcification of rat and human vascular smooth muscle cells in a dose-dependent manner, and this was confirmed by calcium content assay. Similarly, TMAO upregulated the expression of bone-related molecules including Runx2 (Runt-related transcription factor 2) and BMP2 (bone morphogenetic protein-2), suggesting that TMAO promoted osteogenic differentiation of vascular smooth muscle cells. In addition, ex vivo study also showed the positive regulatory effect of TMAO on vascular calcification. Furthermore, we found that TMAO accelerated vascular calcification in rats with chronic kidney disease, as indicated by Mico-computed tomography analysis, alizarin red staining and calcium content assay. By contrast, reducing TMAO levels by antibiotics attenuated vascular calcification in chronic kidney disease rats. Interestingly, TMAO activated NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome and NF-κB (nuclear factor κB) signals during vascular calcification. Inhibition of NLRP3 inflammasome and NF-κB signals attenuated TMAO-induced vascular smooth muscle cell calcification. CONCLUSIONS: This study for the first time demonstrates that TMAO promotes vascular calcification through activation of NLRP3 inflammasome and NF-κB signals, suggesting the potential link between gut microbial metabolism and vascular calcification. Reducing the levels of TMAO could become a potential treatment strategy for vascular calcification in chronic kidney disease.


Asunto(s)
Inflamasomas/efectos de los fármacos , Metilaminas/toxicidad , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Osteogénesis/efectos de los fármacos , Calcificación Vascular/inducido químicamente , Adulto , Anciano , Animales , Antibacterianos/farmacología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Aorta Torácica/patología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamasomas/metabolismo , Masculino , Metilaminas/metabolismo , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , Arterias Tibiales/efectos de los fármacos , Arterias Tibiales/metabolismo , Arterias Tibiales/patología , Técnicas de Cultivo de Tejidos , Calcificación Vascular/metabolismo , Calcificación Vascular/patología , Calcificación Vascular/prevención & control
7.
J Pathol ; 249(4): 461-471, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31397884

RESUMEN

Vascular calcification is prevalent in patients with chronic kidney disease (CKD) and a major risk factor of cardiovascular disease. Vascular calcification is now recognised as a biological process similar to bone formation involving osteogenic differentiation of vascular smooth muscle cells (VSMCs). Cell division cycle 42 (CDC42), a Rac1 family member GTPase, is essential for cartilage development during endochondral bone formation. However, whether CDC42 affects osteogenic differentiation of VSMCs and vascular calcification remains unknown. In the present study, we observed a significant increase in the expression of CDC42 both in rat VSMCs and in calcified arteries during vascular calcification. Alizarin red staining and calcium content assay revealed that adenovirus-mediated CDC42 overexpression led to an apparent VSMC calcification in the presence of calcifying medium, accompanied with up-regulation of bone-related molecules including RUNX2 and BMP2. By contrast, inhibition of CDC42 by ML141 significantly blocked calcification of VSMCs in vitro and aortic rings ex vivo. Moreover, ML141 markedly attenuated vascular calcification in rats with CKD. Furthermore, pharmacological inhibition of AKT signal was shown to block CDC42-induced VSMC calcification. These findings demonstrate for the first time that CDC42 contributes to vascular calcification through a mechanism involving AKT signalling; this uncovered a new function of CDC42 in regulating vascular calcification. This may provide a potential therapeutic target for the treatment of vascular calcification in the context of CKD. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Osteogénesis , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/etiología , Proteína de Unión al GTP cdc42/metabolismo , Anciano , Animales , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/farmacología , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/patología , Transducción de Señal , Sulfonamidas/farmacología , Técnicas de Cultivo de Tejidos , Calcificación Vascular/sangre , Calcificación Vascular/enzimología , Calcificación Vascular/patología
8.
Lab Invest ; 98(10): 1320-1332, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29785051

RESUMEN

Vascular calcification is a highly regulated biological process similar to bone formation involving osteogenic differentiation of vascular smooth muscle cells (VSMCs). Hyaluronan (HA), a major structural component of the extracellular matrix in cartilage, has been shown to inhibit osteoblast differentiation. However, whether HA affects osteogenic differentiation and calcification of VSMCs remains unclear. In the present study, we used in vitro and ex vivo models of vascular calcification to investigate the role of HA in vascular calcification. Both high and low molecular weight HA treatment significantly reduced calcification of rat VSMCs in a dose-dependent manner, as detected by alizarin red staining and calcium content assay. Ex vivo study further confirmed the inhibitory effect of HA on vascular calcification. Similarly, HA treatment decreased ALP activity and expression of bone-related molecules including Runx2, BMP2 and Msx2. By contrast, inhibition of HA synthesis by 4-methylumbelliferone (4MU) promoted calcification of rat VSMCs. In addition, adenovirus-mediated overexpression of HA synthase 2 (HAS2), a major HA synthase in VSMCs, also inhibited calcification of VSMCs, whereas CRISPR/Cas9-mediated HAS2 knockout promoted calcification of rat A10 cells. Furthermore, we found that BMP2 signaling was inhibited in VSMCs after HA treatment. Recombinant BMP2 enhanced high calcium and phosphate-induced VSMC calcification, which can be blocked by HA treatment. Taken together, these findings suggest that HA inhibits vascular calcification involving BMP2 signaling.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Ácido Hialurónico/metabolismo , Calcificación Vascular/etiología , Animales , Línea Celular , Técnicas de Inactivación de Genes , Hialuronano Sintasas/genética , Hialuronano Sintasas/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Cultivo Primario de Células , Ratas Sprague-Dawley , Calcificación Vascular/metabolismo
9.
J Cardiovasc Pharmacol ; 69(6): 382-388, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28581447

RESUMEN

Cold cardioplegia is used to induce heart arrest during cardiac surgery. However, endothelial function may be compromised after this procedure. Accordingly, interventions such as adenosine, that mimic the effects of preconditioning, may minimize endothelial injury. Herein, we investigated whether adenosine prevents cold-induced injury to the endothelium. Cultured human cardiac microvascular endothelial cells were treated with adenosine for different durations. Phosphorylation and expression of endothelial nitric oxide synthase (eNOS), p38MAPK, ERK1/2, and p70S6K6 were measured along with nitric oxide (NO) production using diaminofluorescein-2 diacetate (DAF-2DA) probe. Cold-induced injury by hypothermia to 4°C for 45 minutes to mimic conditions of cold cardioplegia during open heart surgery was induced in human cardiac microvascular endothelial cells. Under basal conditions, adenosine stimulated NO production, eNOS phosphorylation at serine 1177 from 5 minutes to 4 hours and inhibited eNOS phosphorylation at threonine 495 from 5 minutes to 6 hours, but increased phosphorylation of ERK1/2, p38MAPK, and p70S6K only after exposure for 5 minutes. Cold-induced injury inhibited NO production and the phosphorylation of the different enzymes. Importantly, adenosine prevented these effects of hypothermic injury. Our data demonstrated that adenosine prevents hypothermic injury to the endothelium by activating ERK1/2, eNOS, p70S6K, and p38MAPK signaling pathways at early time points. These findings also indicated that 5 minutes after administration of adenosine or release of adenosine is an important time window for cardioprotection during cardiac surgery.


Asunto(s)
Adenosina/administración & dosificación , Frío/efectos adversos , Crioprotectores/administración & dosificación , Células Endoteliales/efectos de los fármacos , Hipotermia Inducida/efectos adversos , Lesiones del Sistema Vascular/prevención & control , Células Cultivadas , Citoprotección , Esquema de Medicación , Células Endoteliales/enzimología , Células Endoteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Lesiones del Sistema Vascular/enzimología , Lesiones del Sistema Vascular/etiología , Lesiones del Sistema Vascular/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Mol Cell Biochem ; 420(1-2): 151-60, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27502306

RESUMEN

Vascular calcification has been considered as a biological process resembling bone formation involving osteogenic differentiation. It is a major risk factor for cardiovascular morbidity and mortality. Previous studies have shown the protective effects of curcumin on cardiovascular diseases. However, whether curcumin has effects on osteogenic differentiation and calcification of vascular smooth muscle cells (VSMCs) has not been reported. In the present study, we used an in vitro model of VSMC calcification to investigate the role of curcumin in the progression of rat VSMC calcification. Curcumin treatment significantly reduced calcification of VSMCs in a dose-dependent manner, detected by alizarin red staining and calcium content assay. Similarly, ALP activity and expression of bone-related molecules including Runx2, BMP2, and Osterix were also decreased in VSMCs treated with curcumin. In addition, flow cytometry analysis and caspase-3 activity assay revealed that curcumin treatment significantly suppressed apoptosis of VSMCs, which plays an important role during vascular calcification. Furthermore, we found that pro-apoptotic molecules including p-JNK and Bax were up-regulated in VSMCs treated with calcifying medium, but they were reduced in VSMCs after curcumin treatment. However, curcumin treatment has no effect on expression of NF-κB p65. Taken together, these findings suggest that curcumin attenuates apoptosis and calcification of VSMCs, presumably via inhibition of JNK/Bax signaling pathway.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Curcumina/farmacología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Osteogénesis/efectos de los fármacos , Calcificación Vascular/metabolismo , Animales , Masculino , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Ratas , Ratas Sprague-Dawley , Calcificación Vascular/patología
11.
Yao Xue Xue Bao ; 51(8): 1281-4, 2016 08.
Artículo en Zh | MEDLINE | ID: mdl-29905994

RESUMEN

This study was conducted to investigate the chemical constituents in the root of Dysosma versipellis(Hance) M. Cheng. The constituents were isolated by silica gel, lichroprep RP-C(18) and pharmadex LH-20 column chromatography and the IR, MS, NMR, 2D-NMR spectroscopic analysis were employed for the structural elucidation. Ten compounds were isolated from the 95% ethanol extract of Dysosma versipellis, their structures were elucidated as dysoverine D (1), dysoverine F (2), dysoverine A (3), podoverine A (4), α-peltatin (5), rutin (6), kaempferol-3-O-ß-D-glucopyranoside (7), quercetin-3-O-ß-D-glucopyranoside (8), kaempferol (9) and quercetin (10). Compound 2 is a new compound, and compounds 1 and 3-6 were isolated from this plant for the first time.


Asunto(s)
Berberidaceae/química , Flavonas/química , Flavonas/aislamiento & purificación , Quempferoles , Podofilotoxina/análogos & derivados , Quercetina , Rutina
13.
Eur J Pharmacol ; 958: 176070, 2023 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-37739306

RESUMEN

Vascular calcification (VC) is highly prevalent and increases the morbidity and mortality of cardiovascular diseases. However, the underlying mechanism remains unclear and there is no effective treatment so far. Interestingly, using systems pharmacology approach, we have predicted that Wogonin (Wog) exhibited potential activity against VC. Then we validated the effect of Wog on VC using human and rat vascular smooth muscle cells (VSMCs), rat arterial rings and vitamin D3-overloaded mouse models. Our results showed that Wog dose-dependently inhibited calcification of VSMCs and rat arterial rings. Consistently, alizarin red staining and calcium content assay confirmed that Wog inhibited aortic calcification in vitamin D3-overloaded mice. Moreover, by constructing the protein regulating network of Wog in suppressing VC, we found heme oxygenase-1 (HMOX-1) was regulated by Wog. Additionally, pathway enrichment analysis revealed that inhibition of reactive oxygen species (ROS) pathway participated in the inhibitory role of Wog in VC and HMOX-1 was also involved in this process. Notably, our study revealed that Wog treatment promoted HMOX-1 expression, and reduced ROS levels in VSMCs. Interestingly, both inhibition of HMOX-1 by ZnPP9 and knockdown of HMOX-1 by siRNA independently eliminated the inhibitory effect of Wog on VC. Finally, administration of Wog suppressed aortic calcification in vitamin D3-overloaded mice and this effect was counteracted by ZnPP9,suggesting the crucial role of HMOX-1 in the inhibitory effect of Wog on VC. Collectively, this study combines systems pharmacology-based strategy and experiments to identify the therapeutic potential of Wog for VC via upregulating HMOX-1 and reducing oxidative stress.

14.
Cardiovasc Res ; 119(13): 2368-2381, 2023 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-37523743

RESUMEN

AIMS: Vascular calcification (VC) is prevalent in pathological processes such as diabetes, chronic kidney disease (CKD), and atherosclerosis, but effective therapies are still lacking by far. Canagliflozin (CANA), a sodium-glucose cotransporter 2 inhibitor, has been approved for the treatment of type 2 diabetes mellitus and exhibits beneficial effects against cardiovascular disease. However, the effect of CANA on VC remains unknown. In this study, we hypothesize that CANA protects against VC. METHODS AND RESULTS: Micro-computed tomography analysis and alizarin red staining revealed that CANA treatment prevented aortic calcification in CKD rats and in VitD3-overloaded mice. Moreover, CANA alleviated the calcification of rat and human arterial rings. Alizarin red staining revealed that calcification of rat and human vascular smooth muscle cells (VSMCs) was attenuated by CANA treatment and this phenomenon was confirmed by calcium content assay. In addition, CANA downregulated the expression of osteogenic differentiation markers Runx2 and BMP2. Of interest, qPCR and western blot analysis revealed that CANA downregulated the expression of the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3), and the downstream signalling molecules Caspase-1 and IL-1ß in VSMCs as well. Both NLRP3 inhibitor MCC950 and knockdown of NLRP3 by siRNA independently resulted in decreased calcification of VSMCs. By contrast, activation of NLRP3 exacerbated VSMC calcification, and this effect was prevented by the addition of CANA. CONCLUSIONS: Our study for the first time demonstrates that CANA exerts a protective effect on VC at least partially via suppressing the NLRP3 signalling pathway. Therefore, supplementation of CANA as well as inhibition of NLRP3 inflammasome presents a potential therapy for VC.


Asunto(s)
Diabetes Mellitus Tipo 2 , Insuficiencia Renal Crónica , Calcificación Vascular , Ratas , Humanos , Ratones , Animales , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Canagliflozina/farmacología , Leucina/metabolismo , Leucina/farmacología , Osteogénesis , Diabetes Mellitus Tipo 2/metabolismo , Dominio Pirina , Microtomografía por Rayos X , Calcificación Vascular/tratamiento farmacológico , Calcificación Vascular/genética , Calcificación Vascular/prevención & control , Insuficiencia Renal Crónica/metabolismo , Glucosa/metabolismo , Nucleótidos/metabolismo , Nucleótidos/farmacología , Sodio/metabolismo , Miocitos del Músculo Liso/metabolismo
15.
Front Pharmacol ; 12: 690392, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335257

RESUMEN

Background: Extensive studies related to vascular calcification (VC) were conducted in recent years. However, no bibliometric analysis has systematically investigated this topic. Our study aimed to determine the hotspots and frontiers of VC research in the past decade and provide a reference for future scientific research directions and decision-making in the VC field. Methods: VC studies were acquired from the Web of Science Core Collection. Bibliometric and visual analyses were performed using CiteSpace, VOSviewer, and Microsoft Excel software. Results: A total of 8,238 English articles on VC research published in 2011-2020 were obtained. In the past decade, annual publications and citations showed a significant growth trend, especially in 2018-2020. The most productive country, institution, journal and author are the United States, the University of California System, PLOS ONE, and Budoff MJ, respectively. The most frequently cited country, journal, and author are the United States, Journal of the American College of Cardiology, and Floege J, respectively. "Vascular calcification," "atherosclerosis," "chronic kidney disease," and "cardiovascular disease" are the primary keywords. The burst keywords "revascularization," "calciprotein particle," "microRNA," and "microcalcification" are speculated to be the research frontiers. Conclusion: The main research hotspots in the VC field are the molecular mechanisms and prognosis of VC in patients with chronic kidney disease or cardiovascular disease. In addition, endovascular therapy and the development of new drugs targeting signal pathways for VC will become the focus of future research. Moreover, non-coding RNAs related to the diagnosis and treatment of VC are great research prospects.

16.
Aging Cell ; 20(6): e13377, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33969611

RESUMEN

Vascular calcification is a common pathologic condition in patients with chronic kidney disease (CKD) and aging individuals. It has been established that vascular calcification is a gene-regulated biological process resembling osteogenesis involving osteogenic differentiation. However, there is no efficient treatment available for vascular calcification so far. The natural polyamine spermidine has been demonstrated to increase life span and protect against cardiovascular disease. It is unclear whether spermidine supplementation inhibits vascular calcification in CKD. Alizarin red staining and quantification of calcium content showed that spermidine treatment markedly reduced mineral deposition in both rat and human vascular smooth muscle cells (VSMCs) under osteogenic conditions. Additionally, western blot analysis revealed that spermidine treatment inhibited osteogenic differentiation of rat and human VSMCs. Moreover, spermidine treatment remarkably attenuated calcification of rat and human arterial rings ex vivo and aortic calcification in rats with CKD. Furthermore, treatment with spermidine induced the upregulation of Sirtuin 1 (SIRT1) in VSMCs and resulted in the downregulation of endoplasmic reticulum (ER) stress signaling components, such as activating transcription factor 4 (ATF4) and CCAAT/enhancer-binding protein homologous protein (CHOP). Both pharmacological inhibition of SIRT1 by SIRT1 inhibitor EX527 and knockdown of SIRT1 by siRNA markedly blocked the inhibitory effect of spermidine on VSMC calcification. Consistently, EX527 abrogated the inhibitory effect of spermidine on aortic calcification in CKD rats. We for the first time demonstrate that spermidine alleviates vascular calcification in CKD by upregulating SIRT1 and inhibiting ER stress, and this may develop a promising therapeutic treatment to ameliorate vascular calcification in CKD.


Asunto(s)
Insuficiencia Renal Crónica/tratamiento farmacológico , Espermidina/uso terapéutico , Calcificación Vascular/tratamiento farmacológico , Animales , Humanos , Masculino , Ratas , Transducción de Señal , Sirtuina 1/metabolismo , Espermidina/farmacología
17.
Free Radic Biol Med ; 172: 530-540, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34174395

RESUMEN

Vascular calcification is very commonly observed in patients with chronic kidney disease (CKD), but there is no efficient therapy available. Oxidative stress plays critical roles in the progression of vascular calcification. Celastrol (Cel), a natural constituent derived from Chinese herbals, exhibits anti-oxidative stress activity. Here, we investigated the effect of celastrol on vascular calcification using vascular smooth muscle cells (VSMCs), arterial rings and CKD rats. Alizarin red staining and gene expression analysis showed that Cel dose-dependently inhibited rat VSMC calcification and osteogenic differentiation. Similarly, ex vivo study revealed that Cel inhibited calcification of rat and human arterial rings. In addition, micro-computed tomography, alizarin red staining and calcium content analysis confirmed that Cel inhibited aortic calcification in CKD rats. Interestingly, Cel treatment increased the mRNA and protein levels of heme oxygenase-1 (HMOX-1), and reduced the levels of reactive oxygen species (ROS) in VSMCs. Furthermore, both pharmacological inhibition of HMOX-1 and knockdown of HMOX-1 by siRNA independently counteracted the inhibitory effect of Cel on vascular calcification. Moreover, knockdown of HMOX-1 prevented Cel treatment-mediated reduction in ROS levels. Finally, Cel treatment reduced Vitamin D3-induced aortic calcification in mice and this effect was blocked by HMOX-1 inhibitor ZnPP9. Collectively, our results suggest that up-regulation of HMOX-1 is required for the inhibitory effect of Cel on vascular calcification. Modulation of HMOX-1 may provide a novel strategy for the treatment of vascular calcification in CKD.


Asunto(s)
Insuficiencia Renal Crónica , Calcificación Vascular , Animales , Células Cultivadas , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso , Osteogénesis , Estrés Oxidativo , Triterpenos Pentacíclicos , Ratas , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/metabolismo , Regulación hacia Arriba , Calcificación Vascular/etiología , Calcificación Vascular/genética , Microtomografía por Rayos X
18.
Fitoterapia ; 146: 104701, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32763365

RESUMEN

Phytochemical investigation of an extract of the rhizome of Curcuma longa L., resulted in the identification of four undescribed bisabolane sesquiterpenoids, namely as bisacurone D-G (1-4). With the aid of comprehensive spectroscopic techniques (NMR, IR, UV, MS), the structures of all isolated compounds were elucidated and subsequently screened for both anti-inflammatory and cytotoxic biological activities, Compounds 1 and 2 showed moderate inhibitory activity toward LPS-induced NO production on RAW 264.7 macrophages.


Asunto(s)
Curcuma/química , Sesquiterpenos Monocíclicos/farmacología , Rizoma/química , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/farmacología , Línea Celular Tumoral , China , Ciclohexanoles , Humanos , Ratones , Estructura Molecular , Sesquiterpenos Monocíclicos/aislamiento & purificación , Óxido Nítrico/metabolismo , Fitoquímicos/aislamiento & purificación , Fitoquímicos/farmacología , Extractos Vegetales/química , Células RAW 264.7 , Sesquiterpenos
19.
Eur J Pharmacol ; 880: 173165, 2020 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-32423869

RESUMEN

Vascular calcification is a highly regulated process similar to osteogenesis involving phenotypic change of vascular smooth muscle cells (VSMCs). 25-Hydroxycholesterol (25-HC), one of oxysterols synthesized by the enzyme cholesterol 25-hydroxylase, has been shown to promote bovine calcifying vascular cells (CVC) calcification. However, whether and how 25-HC regulates vascular calcification are not completely understood. In this study, in vitro and ex vivo models of vascular calcification were used to determine whether 25-HC regulates vascular calcification. Alizarin red staining and calcium content assay showed that 25-HC treatment promoted calcification of rat and human VSMCs in a dose-dependent manner. Similarly, ex vivo study further confirmed that 25-HC accelerated calcification of rat aortic rings. In addition, western blot analysis showed that 25-HC significantly up-regulated the expression of endoplasmic reticulum stress (ERS) signaling molecules including ATF4 and CHOP in VSMCs and flow cytometry analysis revealed that 25-HC increased apoptosis of VSMCs. Moreover, knockdown of CHOP by siRNA blocked 25-HC-induced mineral deposition in VSMCs. Collectively, this study for the first time demonstrates that 25-HC promotes vascular calcification via ATF4/CHOP signaling using in vitro and ex vivo models, suggesting that ERS is involved in the regulation of 25-HC-induced vascular calcification.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Hidroxicolesteroles/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Calcificación Vascular/inducido químicamente , Factor de Transcripción Activador 4/metabolismo , Animales , Aorta Torácica/citología , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Células Cultivadas , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , ARN Interferente Pequeño/genética , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factor de Transcripción CHOP/genética , Calcificación Vascular/metabolismo
20.
Toxicology ; 247(2-3): 133-8, 2008 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-18400355

RESUMEN

We had reported that increased levels of endogenous ghrelin during the progression of doxorubicin-induced cardiomyopathy and heart failure might provide a compensatory self-protective effect. We investigated which pathway(s) produced these protective effects in vitro. Primary cultured cardiomyocytes were induced with doxorubicin in the presence or absence of ghrelin or a tumor necrosis factor-alpha (TNF-alpha) antagonist (etanercept). Ghrelin up-regulated TNF-alpha in a time- and dose-dependent manner. It significantly reduced cell apoptosis and markers of oxidative stress, such as malondialdehyde (MDA) content and lactate dehydrogenase (LDH) activity; it also increased anti-oxidative enzyme activity such as superoxide dismutase (MnSOD) and catalase (CAT), retained mitochondrial membrane potential and energy metabolism compared with doxorubicin alone. Moreover, ghrelin increased mitochondrial anti-apoptosis related gene protein expression such as bcl-2 and MnSOD, reduced cytoplasmic cytochrome C (Cyt C) release and strengthened the activation of NF-kappaB. All these effects were abrogated by etanercept. This suggests ghrelin affects the TNF-alpha/NF-kappaB activation pathways, up-regulating TNF-alpha, to produce anti-oxidative and anti-apoptotic effects that protected cardiomyocytes from doxorubicin-induced cytotoxicity.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Ghrelina/farmacología , Corazón/efectos de los fármacos , Mitocondrias/efectos de los fármacos , FN-kappa B/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA