Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Medicinas Complementares
Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Int Med Res ; 52(3): 3000605241234567, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38530015

RESUMO

OBJECTIVE: Vascular calcification is a common chronic kidney disease complication. This study aimed to investigate the function of long non-coding RNA (LncRNA) H19 in vascular calcification to explore new therapeutic strategies. METHODS: We induced osteogenic differentiation and calcification of vascular smooth muscle cells (VSMCs) using ß-glycerophosphate. Then, we detected the LncRNA H19 promoter methylation status and Erk1/2 pathways using methylation-specific polymerase chain reaction and western blotting, respectively. RESULTS: Compared with the control group, high phosphorus levels induced VSMC calcification, accompanied by increases in LncRNA H19 and the osteogenic marker Runx2 and reduction of the contractile phenotype marker SM22a. LncRNA H19 knockdown inhibited osteogenic differentiation and calcification of VSMCs. However, the suppressed role of VSMC calcification caused by shRNA H19 was partially reversed by simultaneous activation of the Erk1/2 pathways. Mechanically, we found that the methylation rate of CpG islands in the LncRNA H19 promoter region was significantly lower in the high-phosphorus group, and the hypomethylation state elevated LncRNA H19 levels, which in turn regulated phosphorylated Erk1/2 expression. CONCLUSIONS: LncRNA H19 promoted osteogenic differentiation and calcification of VSMCs by regulating the Erk1/2 pathways. Additionally, hypomethylation of LncRNA H19 promoter CpG islands upregulated LncRNA H19 levels and subsequently activated Erk1/2 phosphorylation.


Assuntos
RNA Longo não Codificante , Calcificação Vascular , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Músculo Liso Vascular , Osteogênese/genética , Calcificação Vascular/genética , Calcificação Vascular/metabolismo , Regiões Promotoras Genéticas , Fósforo , Miócitos de Músculo Liso , Células Cultivadas
2.
Cardiovasc Res ; 119(13): 2368-2381, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37523743

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 2 , Insuficiência Renal Crônica , Calcificação Vascular , Ratos , Humanos , Camundongos , Animais , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Canagliflozina/farmacologia , Leucina/metabolismo , Leucina/farmacologia , Osteogênese , Diabetes Mellitus Tipo 2/metabolismo , Domínio Pirina , Microtomografia por Raio-X , Calcificação Vascular/tratamento farmacológico , Calcificação Vascular/genética , Calcificação Vascular/prevenção & controle , Insuficiência Renal Crônica/metabolismo , Glucose/metabolismo , Nucleotídeos/metabolismo , Nucleotídeos/farmacologia , Sódio/metabolismo , Miócitos de Músculo Liso/metabolismo
3.
Nutrients ; 15(13)2023 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-37447244

RESUMO

Phosphorus is a vital element for life found in most foods as a natural component, but it is also one of the most used preservatives added during food processing. High serum phosphorus contributes to develop vascular calcification in chronic kidney disease; however, it is not clear its effect in a population without kidney damage. The objective of this in vivo and in vitro study was to investigate the effect of high phosphorus exposure on the aortic and serum levels of miR-145 and its effect on vascular smooth muscle cell (VSMCs) changes towards less contractile phenotypes. The study was performed in aortas and serum from rats fed standard and high-phosphorus diets, and in VSMCs exposed to different concentrations of phosphorus. In addition, miR-145 silencing and overexpression experiments were carried out. In vivo results showed that in rats with normal renal function fed a high P diet, a significant increase in serum phosphorus was observed which was associated to a significant decrease in the aortic α-actin expression which paralleled the decrease in aortic and serum miR-145 levels, with no changes in the osteogenic markers. In vitro results using VSMCs corroborated the in vivo findings. High phosphorus first reduced miR-145, and afterwards α-actin expression. The miR-145 overexpression significantly increased α-actin expression and partially prevented the increase in calcium content. These results suggest that miR-145 could be an early biomarker of vascular calcification, which could give information about the initiation of the transdifferentiation process in VSMCs.


Assuntos
MicroRNAs , Calcificação Vascular , Ratos , Animais , Fósforo/metabolismo , Músculo Liso Vascular , Actinas/metabolismo , Transdiferenciação Celular , MicroRNAs/genética , MicroRNAs/metabolismo , Fenótipo , Calcificação Vascular/genética , Calcificação Vascular/metabolismo , Miócitos de Músculo Liso , Células Cultivadas
4.
Cells ; 12(4)2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36831311

RESUMO

Increased senescent cell burden and dysregulation of the nuclear factor erythroid 2-related factor 2 (NRF2) pathway have been associated with numerous age-related pathologies; however, their role in promoting vascular calcification (VC) in chronic kidney disease (CKD) has yet to be determined. We investigated whether senescence and NRF2 pathways may serve as drivers of uremia-induced VC using three complementary approaches: a novel model of induced VC in 5/6-nephrectomized rats supplemented with high phosphate and vitamin D; epigastric arteries from CKD patients with established medial calcification; and vascular smooth muscle cells (VSMCs) incubated with uremic serum. Expression of p16Ink4a and p21Cip1, as well as γ-H2A-positive cells, confirmed increased senescent cell burden at the site of calcium deposits in aortic sections in rats, and was similarly observed in calcified epigastric arteries from CKD patients through increased p16Ink4a expression. However, uremic serum-induced VSMC calcification was not accompanied by senescence. Expression of NRF2 and downstream genes, Nqo1 and Sod1, was associated with calcification in uremic rats, while no difference was observed between calcified and non-calcified EAs. Conversely, in vitro uremic serum-driven VC was associated with depleted NRF2 expression. Together, our data strengthen the importance of senescence and NRF2 pathways as potential therapeutic options to combat VC in CKD.


Assuntos
Insuficiência Renal Crônica , Calcificação Vascular , Ratos , Animais , Fator 2 Relacionado a NF-E2/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Músculo Liso Vascular/metabolismo , Calcificação Vascular/genética , Insuficiência Renal Crônica/patologia , Senescência Celular
5.
Nephrol Dial Transplant ; 38(7): 1729-1740, 2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-36722155

RESUMO

INTRODUCTION: MicroRNAs (miRs) regulate vascular calcification (VC), and their quantification may contribute to suspicion of the presence of VC. METHODS: The study was performed in four phases. Phase 1: miRs sequencing of rat calcified and non-calcified aortas. Phase 2: miRs with the highest rate of change, plus miR-145 [the most abundant miR in vascular smooth muscle cells (VSMCs)], were validated in aortas and serum from rats with and without VC. Phase 3: the selected miRs were analyzed in epigastric arteries from kidney donors and recipients, and serum samples from general population. Phase 4: VSMCs were exposed to different phosphorus concentrations, and miR-145 and miR-486 were overexpressed to investigate their role in VC. RESULTS: miR-145, miR-122-5p, miR-486 and miR-598-3p decreased in the rat calcified aortas, but only miR-145 and miR-486 were detected in serum. In human epigastric arteries, miR-145 and miR-486 were lower in kidney transplant recipients compared with donors. Both miRs inversely correlated with arterial calcium content and with VC (Kauppila index). In the general population, the severe VC was associated with the lowest serum levels of both miRs. The receiver operating characteristic curve showed that serum miR-145 was a good biomarker of VC. In VSMCs exposed to high phosphorus, calcium content, osteogenic markers (Runx2 and Osterix) increased, and the contractile marker (α-actin), miR-145 and miR-486 decreased. Overexpression of miR-145, and to a lesser extent miR-486, prevented the increase in calcium content induced by high phosphorus, the osteogenic differentiation and the loss of the contractile phenotype. CONCLUSION: miR-145 and miR-486 regulate the osteogenic differentiation of VSMCs, and their quantification in serum could serve as a marker of VC.


Assuntos
MicroRNAs , Calcificação Vascular , Animais , Humanos , Ratos , Biomarcadores , Cálcio , MicroRNAs/genética , Músculo Liso Vascular , Miócitos de Músculo Liso , Osteogênese/genética , Fósforo , Calcificação Vascular/genética
6.
Kidney Int ; 102(6): 1259-1275, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36063875

RESUMO

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.


Assuntos
Ferroptose , Insuficiência Renal Crônica , Calcificação Vascular , Humanos , Ratos , Camundongos , Animais , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Músculo Liso Vascular , Osteogênese , Cálcio/metabolismo , Antiporters/metabolismo , Miócitos de Músculo Liso/metabolismo , Calcificação Vascular/genética , Calcificação Vascular/prevenção & controle , Ferro/metabolismo , Glutationa/metabolismo , Insuficiência Renal Crônica/patologia , Fosfatos/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo
7.
J Pathol ; 258(3): 213-226, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35894849

RESUMO

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.


Assuntos
Insuficiência Renal Crônica , Calcificação Vascular , Ácido 3-Hidroxibutírico/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Regulação para Baixo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Cetonas/metabolismo , Camundongos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/patologia , NF-kappa B/metabolismo , Ratos , Insuficiência Renal Crônica/patologia , Proteínas Repressoras/metabolismo , Calcificação Vascular/genética , Calcificação Vascular/prevenção & controle
8.
Cell Cycle ; 21(16): 1667-1683, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35435133

RESUMO

Vascular calcification, characterized by the accumulation of calcium-phosphate crystals in blood vessels, is a major cause of cardiovascular complications and chronic kidney disease (CKD)-related death. This work focuses on the molecules involved in high-phosphorus-mediated vascular calcification in CKD. A rat model of CKD was established by 5/6 nephrectomy, and the rats were given normal phosphorus diet (NPD) or high phosphorus diet (HPD). HPD decreased kidney function, increased the concentration of calcium ion and damaged vascular structure in the thoracic aorta of diseased rats. A high phosphorus condition enhanced calcium deposition in vascular smooth muscle cells (VSMCs). High phosphorus also increased the expression of RUNX2 whereas reduced the expression of α-SM actin in the aortic tissues and VSMCs. Long non-coding RNA (lncRNA) H19 was upregulated in the aortic tissues after HPD treatment. H19 bound to microRNA (miR)-138 to block its inhibitory effect on TLR3 mRNA and activated the NF-κB signaling pathway. Downregulation of H19 or TLR3 alleviated, whereas downregulation of miR-138 aggravated the calcification and vascular damage in model rats and VSMCs. In conclusion, this study demonstrates that the H19/miR-138/TLR3 axis is involved in high phosphorus-mediated vascular calcification in rats with CKD.


Assuntos
MicroRNAs , RNA Longo não Codificante , Insuficiência Renal Crônica , Calcificação Vascular , Animais , Cálcio/metabolismo , Células Cultivadas , MicroRNAs/genética , MicroRNAs/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fósforo/metabolismo , Fósforo/farmacologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Ratos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo , Receptor 3 Toll-Like/metabolismo , Calcificação Vascular/genética , Calcificação Vascular/metabolismo
9.
Exp Dermatol ; 31(4): 548-555, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34758173

RESUMO

Pseudoxanthoma elasticum (PXE; OMIM 264800) is a rare heritable multisystem disorder, characterized by ectopic mineralization affecting elastic fibres in the skin, eyes and the cardiovascular system. Skin findings often lead to early diagnosis of PXE, but currently, no specific treatment exists to counteract the progression of symptoms. PXE belongs to a group of Mendelian calcification disorders linked to pyrophosphate metabolism, which also includes generalized arterial calcification of infancy (GACI) and arterial calcification due to CD73 deficiency (ACDC). Inactivating mutations in ABCC6, ENPP1 and NT5E are the genetic cause of these diseases, respectively, and all of them result in reduced inorganic pyrophosphate (PPi ) concentration in the circulation. Although PPi is a strong inhibitor of ectopic calcification, oral supplementation therapy was initially not considered because of its low bioavailability. Our earlier work however demonstrated that orally administered pyrophosphate inhibits ectopic calcification in the animal models of PXE and GACI, and that orally given Na4 P2 O7 is absorbed in humans. Here, we report that gelatin-encapsulated Na2 H2 P2 O7  has similar absorption properties in healthy volunteers and people affected by PXE. The sodium-free K2 H2 P2 O7 form resulted in similar uptake in healthy volunteers and inhibited calcification in Abcc6-/- mice as effectively as its sodium counterpart. Novel pyrophosphate compounds showing higher bioavailability in mice were also identified. Our results provide an important step towards testing oral PPi in clinical trials in PXE, or potentially any condition accompanied by ectopic calcification including diabetes, chronic kidney disease or ageing.


Assuntos
Pseudoxantoma Elástico , Calcificação Vascular , Animais , Suplementos Nutricionais , Difosfatos , Humanos , Camundongos , Mutação , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Diester Fosfórico Hidrolases/uso terapêutico , Pseudoxantoma Elástico/tratamento farmacológico , Pseudoxantoma Elástico/genética , Pseudoxantoma Elástico/metabolismo , Pirofosfatases/genética , Pirofosfatases/metabolismo , Pirofosfatases/uso terapêutico , Calcificação Vascular/tratamento farmacológico , Calcificação Vascular/genética
10.
Aging (Albany NY) ; 13(12): 16749-16762, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34170851

RESUMO

Our previous study showed that bone marrow mesenchymal stem cell derived exosomes (BMSC-Exos) suppress high phosphorus (Pi)-induced calcification of vascular smooth muscle cells (VSMCs). However, the mechanism had remained unclear. This study aimed to investigate the mechanism by which BMSC-Exos inhibit vascular calcification (VC). We found that BMSC-Exos reduced high Pi-induced Runx2, osteocalcin and BMP2 expression and inhibited the calcium deposition. Gene expression of human VSMCs stimulated by Pi or Pi plus BMSC-Exos (Pi + Exo) was systematically examined by microarray technology. NONHSAT 084969.2 and transcription factor p65 expression was significantly lower in the Pi + Exo group compared with the Pi group. This finding indicated that NONHSAT 084969.2 and the nuclear factor-κB pathway might play an important role in VC inhibition by BMSC-Exos. By silencing NONHSAT 084969.2 with small interfering RNA, Runx2, BMP2, and osteocalcin expression was decreased significantly. The calcified nodule content and alkaline phosphatase activity were reduced after NONHSAT 084969.2 inhibition and p65, p50, and IκB kinase-α expression was decreased significantly. These results indicated that BMSC-Exos inhibited Pi-induced transdifferentiation and calcification of VSMCs by regulating the NONHSAT 084969.2/nuclear factor-κB axis.


Assuntos
Exossomos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , NF-kappa B/metabolismo , Fósforo/toxicidade , RNA Longo não Codificante/metabolismo , Calcificação Vascular/genética , Linhagem Celular , Transdiferenciação Celular/efeitos dos fármacos , Análise por Conglomerados , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Exossomos/ultraestrutura , Perfilação da Expressão Gênica , Humanos , Minerais/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , RNA Longo não Codificante/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Calcificação Vascular/patologia
11.
Sci Rep ; 10(1): 1645, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32015399

RESUMO

Arterial medial calcification (AMC) involves an increased small extracellular vesicle (sEV) secretion and apatite calcium precipitation in the arterial wall. The mechanisms mediating AMC remain poorly understood. In the present study, smooth muscle-specific acid ceramidase (Ac) gene knockout mice (Asah1fl/fl/SMCre) were used to demonstrate the role of lysosomal ceramide signaling pathway in AMC. Asah1fl/fl/SMCre mice were found to have more severe AMC in both aorta and coronary arteries compared to their littermates (Asah1fl/fl/SMwt and WT/WT mice) after receiving a high dose vitamin D. These mice also had pronounced upregulation of osteopontin and RUNX2 (osteogenic markers), CD63, AnX2 (sEV markers) and ALP expression (mineralization marker) in the arterial media. In cultured coronary arterial smooth muscle cells (CASMCs) from Asah1fl/fl/SMCre mice, high dose of Pi led to a significantly increased calcium deposition, phenotypic change and sEV secretion compared to WT CASMCs, which was associated with reduced lysosome-multivesicular body (MVB) interaction. Also, GW4869, sEV release inhibitor decreased sEV secretion and calcification in these cells. Lysosomal transient receptor potential mucolipin 1 (TRPML1) channels regulating lysosome interaction with MVBs were found remarkably inhibited in Asah1fl/fl/SMCre CASMCs as shown by GCaMP3 Ca2+ imaging and Port-a-Patch patch clamping of lysosomes. Lysosomal Ac in SMCs controls sEV release by regulating lysosomal TRPML1 channel activity and lysosome-MVB interaction, which importantly contributes to phenotypic transition and AMC.


Assuntos
Ceramidase Ácida/metabolismo , Calcificação Vascular/metabolismo , Ceramidase Ácida/genética , Animais , Aorta/metabolismo , Aorta/patologia , Sinalização do Cálcio , Células Cultivadas , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Modelos Animais de Doenças , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patologia , Lipogranulomatose de Farber/genética , Lipogranulomatose de Farber/metabolismo , Lisossomos/metabolismo , Masculino , Redes e Vias Metabólicas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Cardiovasculares , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Esfingolipídeos/metabolismo , Canais de Potencial de Receptor Transitório/agonistas , Canais de Potencial de Receptor Transitório/metabolismo , Calcificação Vascular/genética , Calcificação Vascular/patologia
12.
Kidney Int ; 97(3): 487-501, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31866113

RESUMO

Klotho knock-out mice are an important model for vascular calcification, which is associated with chronic kidney disease. In chronic kidney disease, serum magnesium inversely correlates with vascular calcification. Here we determine the effects of serum magnesium on aortic calcification in Klotho knock-out mice treated with a minimal or a high magnesium diet from birth. After eight weeks, serum biochemistry and aorta and bone tissues were studied. Protective effects of magnesium were characterized by RNA-sequencing of the aorta and micro-CT analysis was performed to study bone integrity. A high magnesium diet prevented vascular calcification and aortic gene expression of Runx2 and matrix Gla protein found in such mice on the minimal magnesium diet. Differential expression of inflammation and extracellular matrix remodeling genes accompanied the beneficial effects of magnesium on calcification. High dietary magnesium did not affect serum parathyroid hormone, 1,25-dihydroxyvitamin D3 or calcium. High magnesium intake prevented vascular calcification despite increased fibroblast growth factor-23 and phosphate concentration in the knock-out mice. Compared to mice on the minimal magnesium diet, the high magnesium diet reduced femoral bone mineral density by 20% and caused excessive osteoid formation indicating osteomalacia. Osteoclast activity was unaffected by the high magnesium diet. In Saos-2 osteoblasts, magnesium supplementation reduced mineralization independent of osteoblast function. Thus, high dietary magnesium prevents calcification in Klotho knock-out mice. These effects are potentially mediated by reduction of inflammatory and extracellular matrix remodeling pathways within the aorta. Hence magnesium treatment may be promising to prevent vascular calcification, but the risk for osteomalacia should be considered.


Assuntos
Glucuronidase/deficiência , Magnésio/farmacologia , Insuficiência Renal Crônica , Calcificação Vascular , Animais , Glucuronidase/genética , Proteínas Klotho , Camundongos , Camundongos Knockout , Hormônio Paratireóideo , Fosfatos , Calcificação Vascular/genética , Calcificação Vascular/prevenção & controle
13.
Curr Opin Lipidol ; 30(5): 357-363, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31369409

RESUMO

PURPOSE OF REVIEW: This review addresses recent developments in studies of lipid regulation of calcific disease of arteries and cardiac valves, including the role of nuclear receptors. The role of lipid-soluble signals and their receptors is timely given the recent evidence and concerns that lipid-lowering treatment may increase the rate of progression of coronary artery calcification, which has been long associated with increased cardiovascular risk. Understanding the mechanisms will be important for interpreting such clinical information. RECENT FINDINGS: New findings support regulation of calcific vascular and valvular disease by nuclear receptors, including the vitamin D receptor, glucocorticoid receptor, nutrient-sensing nuclear receptors (liver X receptor, farnesoid X receptor, and peroxisome proliferator-activated receptors), and sex hormone (estrogen and androgen) receptors. There were two major unexpected findings: first, vitamin D supplementation, which was previously believed to prevent or reduce vascular calcification, showed no cardiovascular benefit in large randomized, controlled trials. Second, both epidemiological studies and coronary intravascular ultrasound studies suggest that treatment with HMG-CoA reductase inhibitors increases progression of coronary artery calcification, raising a question of whether there are mechanically stable and unstable forms of coronary calcification. SUMMARY: For clinical practice and research, these new findings offer new fundamental mechanisms for vascular calcification and provide new cautionary insights for therapeutic avenues.


Assuntos
Calcinose/genética , Doenças das Valvas Cardíacas/genética , Receptores Citoplasmáticos e Nucleares/genética , Calcificação Vascular/genética , Artérias/efeitos dos fármacos , Artérias/patologia , Calcinose/patologia , Doenças das Valvas Cardíacas/tratamento farmacológico , Doenças das Valvas Cardíacas/patologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Metabolismo dos Lipídeos/genética , Receptores X do Fígado/genética , Receptores Ativados por Proliferador de Peroxissomo/genética , Calcificação Vascular/tratamento farmacológico , Calcificação Vascular/patologia
14.
J Am Heart Assoc ; 8(14): e012486, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31304837

RESUMO

Background Homoarginine ( hA rg) has been shown to be cardioprotective in a model of ischemic heart failure; however, the mechanism remains unknown. hA rg can inhibit tissue-nonspecific alkaline phosphatase ( TNAP ), an enzyme that promotes vascular calcification. We hypothesized that hA rg will exert beneficial effects by reducing calcification in a mouse model of coronary artery disease associated with TNAP overexpression and hypercholesterolemia. Methods and Results TNAP was overexpressed in the endothelium in mice homozygous for a low-density lipoprotein receptor mutation (wicked high cholesterol [ WHC ] allele). WHC and WHC -endothelial TNAP mice received placebo or hA rg supplementation (14 mg/L in drinking water) starting at 6 weeks of age simultaneously with an atherogenic diet. Outcomes were compared between the groups after 4 to 5 weeks on treatment. Experiments were performed in males, which presented a study limitation. As expected, WHC -endothelial TNAP mice on the placebo had increased mortality (median survival 27 days, P<0.0001), increased coronary calcium and lipids ( P<0.01), increased left ventricular end-diastolic diameter ( P<0.0001), reduced ejection fraction ( P<0.05), and increased myocardial fibrosis ( P<0.0001) compared with WHC mice. Contrary to our hypothesis, hA rg neither inhibited TNAP activity in vivo nor reduced coronary artery calcification and atherosclerosis in WHC -endothelial TNAP mice; however, compared with the placebo, hA rg prevented left ventricular dilatation ( P<0.01), preserved ejection fraction ( P<0.05), and reduced myocardial fibrosis ( P<0.001). Conclusions The beneficial effect of hA rg supplementation in the setting of calcified coronary artery disease is likely due to its direct protective actions on the myocardial response to the ischemic injury and not to the inhibition of TNAP activity and calcification.


Assuntos
Fosfatase Alcalina/efeitos dos fármacos , Doença da Artéria Coronariana/fisiopatologia , Endotélio/efeitos dos fármacos , Coração/efeitos dos fármacos , Homoarginina/farmacologia , Calcificação Vascular/patologia , Função Ventricular Esquerda/efeitos dos fármacos , Fosfatase Alcalina/genética , Animais , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/patologia , Dieta Aterogênica , Dilatação Patológica/diagnóstico por imagem , Dilatação Patológica/genética , Dilatação Patológica/fisiopatologia , Modelos Animais de Doenças , Ecocardiografia , Endotélio/metabolismo , Fibrose , Hipercolesterolemia/genética , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Miocárdio/patologia , Receptores de LDL/genética , Volume Sistólico/efeitos dos fármacos , Volume Sistólico/genética , Taxa de Sobrevida , Sístole , Calcificação Vascular/genética , Função Ventricular Esquerda/genética
15.
Funct Integr Genomics ; 19(4): 633-643, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30850904

RESUMO

Vascular calcification is a common complication in patients with chronic kidney disease (CKD). It is an important predictor of cardiovascular disease and all-cause mortality. Previous studies have confirmed that bone marrow mesenchymal stem cell (BMSC) therapy can reduce vascular calcification, but the specific mechanism is still controversial. In this study, we aimed to investigate the mechanisms of BMSC-derived exosomes (EXO) in improving vascular calcification. BMSCs were cultured and EXO were isolated using the Total Exosome Isolation Reagent. Human aortic vascular smooth muscle cells (HA-VSMCs) were cultured into three groups: control group, high phosphorus group, and high phosphorus plus EXO group. Then, indicators related to smooth muscle cell calcification and microRNA profiles were analyzed. BMSC-derived exosomes inhibited high phosphorus-induced calcification in HA-VSMCs. Besides, EXO treatment reduced calcium content and decreased the alkaline phosphatase (AKP) activity in high phosphorus co-incubated HA-VSMCs. MicroRNA (miRNA) and mRNA expression profiles analyses revealed that 63 miRNAs were significantly upregulated and 1424 genes were significantly downregulated in HA-VSMCs after EXO treatment. Functional miRNA-gene regulatory network revealed that mTOR, MAPK, and Wnt signaling pathway were involved in vascular calcification. BMSC-derived exosomes alleviated high phosphorus-induced calcification in HA-VSMC through modifying miRNA profiles.


Assuntos
Exossomos/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Calcificação Vascular/metabolismo , Fosfatase Alcalina/metabolismo , Cálcio/metabolismo , Células Cultivadas , Exossomos/genética , Redes Reguladoras de Genes , Humanos , MicroRNAs/metabolismo , Fósforo/toxicidade , Transcriptoma , Regulação para Cima , Calcificação Vascular/etiologia , Calcificação Vascular/genética
16.
Int J Mol Sci ; 20(3)2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30717170

RESUMO

Matrix Gla Protein (MGP), a small Gla vitamin K-dependent protein, is the most powerful natural occurring inhibitor of calcification in the human body. To become biologically active, MGP must undergo vitamin K-dependent carboxylation and phosphorylation. Vitamin K deficiency leads to the inactive uncarboxylated, dephosphorylated form of MGP (dpucMGP). We aimed to review the existing data on the association between circulating dpucMGP and vascular calcification, renal function, mortality, and cardiovascular disease in distinct populations. Moreover, the association between vitamin K supplementation and serum levels of dpucMGP was also reviewed.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Doenças Cardiovasculares/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Processamento de Proteína Pós-Traducional , Insuficiência Renal Crônica/metabolismo , Calcificação Vascular/metabolismo , Deficiência de Vitamina K/metabolismo , Vitamina K/metabolismo , Transporte Biológico , Proteínas de Ligação ao Cálcio/genética , Doenças Cardiovasculares/complicações , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/mortalidade , Suplementos Nutricionais , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica , Humanos , Fosforilação , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/mortalidade , Análise de Sobrevida , Calcificação Vascular/complicações , Calcificação Vascular/genética , Calcificação Vascular/mortalidade , Rigidez Vascular , Deficiência de Vitamina K/complicações , Deficiência de Vitamina K/genética , Deficiência de Vitamina K/mortalidade , Proteína de Matriz Gla
17.
J Invest Dermatol ; 139(2): 360-368, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30130617

RESUMO

Pseudoxanthoma elasticum (PXE), a prototype of heritable ectopic mineralization disorders, is caused by mutations in the ABCC6 gene encoding a putative efflux transporter ABCC6. It was recently shown that the absence of ABCC6-mediated adenosine triphosphate release from the liver and, consequently, reduced inorganic pyrophosphate levels underlie the pathogenesis of PXE. Given that tissue-nonspecific alkaline phosphatase (TNAP), encoded by ALPL, is the enzyme responsible for degrading inorganic pyrophosphate, we hypothesized that reducing TNAP levels either by genetic or pharmacological means would lead to amelioration of the ectopic mineralization phenotype in the Abcc6-/- mouse model of PXE. Thus, we bred Abcc6-/- mice to heterozygous Alpl+/- mice that display approximately 50% plasma TNAP activity. The Abcc6-/-Alpl+/- double-mutant mice showed 52% reduction of mineralization in the muzzle skin compared with the Abcc6-/-Alpl+/+ mice. Subsequently, oral administration of SBI-425, a small molecule inhibitor of TNAP, resulted in 61% reduction of plasma TNAP activity and 58% reduction of mineralization in the muzzle skin of Abcc6-/- mice. By contrast, SBI-425 treatment of Enpp1 mutant mice, another model of ectopic mineralization associated with reduced inorganic pyrophosphate, failed to reduce muzzle skin mineralization. These results suggest that inhibition of TNAP might provide a promising treatment strategy for PXE, a currently intractable disease.


Assuntos
Niacinamida/análogos & derivados , Pseudoxantoma Elástico/tratamento farmacológico , Pirofosfatases/antagonistas & inibidores , Sulfonamidas/administração & dosagem , Trifosfato de Adenosina/metabolismo , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Difosfatos/sangue , Difosfatos/metabolismo , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Mutação , Niacinamida/administração & dosagem , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Pseudoxantoma Elástico/sangue , Pseudoxantoma Elástico/genética , Pirofosfatases/genética , Pirofosfatases/metabolismo , Pele/metabolismo , Pele/patologia , Calcificação Vascular/sangue , Calcificação Vascular/tratamento farmacológico , Calcificação Vascular/genética
18.
Am J Pathol ; 189(2): 216-225, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30414410

RESUMO

Ectopic mineralization is a global problem and leading cause of morbidity and mortality. The pathomechanisms of ectopic mineralization are poorly understood. Recent studies on heritable ectopic mineralization disorders with defined gene defects have been helpful in elucidation of the mechanisms of ectopic mineralization in general. The prototype of such disorders is pseudoxanthoma elasticum (PXE), a late-onset, slowly progressing disorder with multisystem clinical manifestations. Other conditions include generalized arterial calcification of infancy (GACI), characterized by severe, early-onset mineralization of the cardiovascular system, often with early postnatal demise. In addition, arterial calcification due to CD73 deficiency (ACDC) occurs late in life, mostly affecting arteries in the lower extremities in elderly individuals. These three conditions, PXE, GACI, and ACDC, caused by mutations in ABCC6, ENPP1, and NT5E, respectively, are characterized by reduced levels of inorganic pyrophosphate (PPi) in plasma. Because PPi is a powerful antimineralization factor, it has been postulated that reduced PPi is a major determinant for ectopic mineralization in these conditions. These and related observations on complementary mechanisms of ectopic mineralization have resulted in development of potential treatment modalities for PXE, including administration of bisphosphonates, stable PPi analogs with antimineralization activity. It is conceivable that efficient treatments may soon become available for heritable ectopic mineralization disorders with application to common calcification disorders.


Assuntos
5'-Nucleotidase/deficiência , Difosfonatos/uso terapêutico , Pseudoxantoma Elástico , Calcificação Vascular , Difosfatos/sangue , Proteínas Ligadas por GPI/deficiência , Humanos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Pseudoxantoma Elástico/sangue , Pseudoxantoma Elástico/tratamento farmacológico , Pseudoxantoma Elástico/genética , Pseudoxantoma Elástico/patologia , Pirofosfatases/genética , Pirofosfatases/metabolismo , Calcificação Vascular/sangue , Calcificação Vascular/tratamento farmacológico , Calcificação Vascular/genética , Calcificação Vascular/patologia
19.
Sci Rep ; 8(1): 13733, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30214074

RESUMO

Vitamin K plays a crucial role in the regulation of vascular calcifications by allowing activation of matrix Gla protein. The dietary requirement for vitamin K is low because of an efficient recycling of vitamin K by vitamin K epoxide reductase (VKORC1). However, decreased VKORC1 activity may result in vascular calcification. More than 30 coding mutations of VKORC1 have been described. While these mutations have been suspected of causing anticoagulant resistance, their association with an increase in the risk of vascular calcification has never been considered. We thus investigated functional cardiovascular characteristics in a rat model mutated in VKORC1. This study revealed that limited intake in vitamin K in mutated rat induced massive calcified areas in the media of arteries of lung, aortic arch, kidneys and testis. Development of calcifications could be inhibited by vitamin K supplementation. In calcified areas, inactive Matrix Gla protein expression increased, while corresponding mRNA expression was not modified. Mutation in VKORC1 associated with a limited vitamin K intake is thus a major risk for cardiovascular disease. Our model is the first non-invasive rat model that shows spontaneous medial calcifications and would be useful for studying physiological function of vitamin K.


Assuntos
Arteriosclerose/genética , Esclerose Calcificante da Média de Monckeberg/genética , Calcificação Vascular/genética , Vitamina K Epóxido Redutases/genética , Vitamina K/metabolismo , Animais , Anticoagulantes/administração & dosagem , Aorta Torácica/patologia , Artérias/patologia , Arteriosclerose/patologia , Modelos Animais de Doenças , Humanos , Rim/irrigação sanguínea , Rim/patologia , Pulmão/irrigação sanguínea , Pulmão/patologia , Masculino , Esclerose Calcificante da Média de Monckeberg/patologia , Mutação , Mutação de Sentido Incorreto/genética , Polimorfismo de Nucleotídeo Único/genética , Ratos , Testículo/irrigação sanguínea , Testículo/patologia , Calcificação Vascular/patologia , Vitamina K/genética , Vitamina K Epóxido Redutases/metabolismo
20.
Kidney Int ; 94(4): 716-727, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30041812

RESUMO

PiT-2, a type III sodium-dependent phosphate transporter, is a causative gene for the brain arteriolar calcification in people with familial basal ganglion calcification. Here we examined the effect of PiT-2 haploinsufficiency on vascular calcification in uremic mice using wild-type and global PiT-2 heterozygous knockout mice. PiT-2 haploinsufficiency enhanced the development of vascular calcification in mice with chronic kidney disease fed a high-phosphate diet. No differences were observed in the serum mineral biomarkers and kidney function between the wild-type and PiT-2 heterozygous knockout groups. Micro computed tomography analyses of femurs showed that haploinsufficiency of PiT-2 decreased trabecular bone mineral density in uremia. In vitro, sodium-dependent phosphate uptake was decreased in cultured vascular smooth muscle cells isolated from PiT-2 heterozygous knockout mice compared with those from wild-type mice. PiT-2 haploinsufficiency increased phosphate-induced calcification of cultured vascular smooth muscle cells compared to the wild-type. Furthermore, compared to wild-type vascular smooth muscle cells, PiT-2 deficient vascular smooth muscle cells had lower osteoprotegerin levels and increased matrix calcification, which was attenuated by osteoprotegerin supplementation. Thus, PiT-2 in vascular smooth muscle cells protects against phosphate-induced vascular calcification and may be a therapeutic target in the chronic kidney disease population.


Assuntos
Fosfatos/metabolismo , Insuficiência Renal Crônica/complicações , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Calcificação Vascular/genética , Animais , Biomarcadores/sangue , Densidade Óssea/genética , Feminino , Haploinsuficiência , Heterozigoto , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/metabolismo , Osteoprotegerina/metabolismo , Fosfatos/administração & dosagem , Insuficiência Renal Crônica/sangue , Uremia/complicações , Calcificação Vascular/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA