Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Vascul Pharmacol ; 146: 107096, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35952961

RESUMEN

Recent studies have shown that short-chain fatty acids (SCFAs), primarily acetate, propionate and butyrate, play a crucial role in the pathogenesis of cardiovascular disease. Whether SCFAs regulate vascular calcification, a common pathological change in cardiovascular tissues, remains unclear. This study aimed to investigate the potential role of SCFAs in vascular calcification. Using cellular and animal models of vascular calcification, we showed that butyrate significantly enhanced high phosphate (Pi)-induced calcification and osteogenic transition of vascular smooth muscle cells (VSMC) in vitro, whereas acetate and propionate had no effects. Subsequent studies confirmed that butyrate significantly promoted high Pi-induced aortic ring calcification ex vivo and high dose vitamin D3 (vD3)-induced mouse vascular calcification in vivo. Mechanistically, butyrate significantly inhibited histone deacetylase (HDAC) expression in VSMCs, and a pan HDAC inhibitor Trichostatin A showed similar inductive effects on calcification and osteogenic transition of VSMCs to butyrate. In addition, the SCFA sensing receptors Gpr41 and Gpr109a were primarily expressed by VSMCs, and butyrate induced the rapid activation of NF-κB, Wnt and Akt signaling in VSMCs. Intriguingly, the NF-κB inhibitor SC75741 significantly attenuated butyrate-induced calcification and the osteogenic gene Msx2 expression in VSMCs. We showed that knockdown of Gpr41 but not Gpr109a attenuated butyrate-induced VSMC calcification. This study reveals that butyrate accelerates vascular calcification via its dual effects on HDAC inhibition and NF-κB activation. Our data provide novel insights into the role of microbe-host interaction in vascular calcification, and may have implications for the development of potential therapy for vascular calcification.


Asunto(s)
FN-kappa B , Calcificación Vascular , Animales , Butiratos/metabolismo , Butiratos/farmacología , Células Cultivadas , Inhibidores de Histona Desacetilasas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Fosfatos , Propionatos/metabolismo , Propionatos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Calcificación Vascular/patología , Vitamina D
2.
Bioengineered ; 13(3): 5277-5291, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35168476

RESUMEN

The current research aimed to verify the effects of erythropoietin (EPO) on vascular calcification under inflammatory conditions and the molecular regulator of vascular calcification induced by EPO. To induce vascular calcification and systemic chronic inflammation in SD rats, EPO was administered intraperitoneally, and 10% casein was injected subcutaneously. The administration period lasted for 20 consecutive weeks. Blood samples were subsequently collected to detect inflammatory factors and vascular calcification. Additionally, high-dose EPOs were applied to stimulate primary vascular smooth muscle cells (VSMCs), and vascular calcification was measured using alizarin red staining, alkaline phosphatase (ALP) activity, and calcium salt quantification. The probe 2',7'-dichlorofluorescein diacetate (DCFH-DA) was employed to detect cellular reactive oxygen species (ROS) levels. The expressions of bone formation-related protein and anti-calcification protein matrix gla protein (MGP) were determined via Western blot. Compared with the control group, calcium deposits and vascular calcification were increased in the EPO group, tumor necrosis factor-alpha (TNF-α) group and TNF-α+ EPO group, whereas MGP was significantly reduced. Moreover, under the stimulation of TNF-α and EPO+TNF-α, pp38/p38 was increased substantially, the addition of p38 inhibitor SB203580 could significantly reduce calcium deposits and vascular calcification. In vivo experiment, compared with the EPO group, calcium salt deposition and vascular calcification were elevated in the EPO+casein group. The present results revealed that high-dose EPO could cause calcification of the abdominal aorta in rats. The inflammatory response aggravated the vascular calcification induced by EPO via activating p38 and ROS levels.


Asunto(s)
Eritropoyetina , Calcificación Vascular , Animales , Calcio/metabolismo , Caseínas/efectos adversos , Caseínas/metabolismo , Células Cultivadas , Eritropoyetina/efectos adversos , Eritropoyetina/metabolismo , Inflamación/metabolismo , Músculo Liso Vascular/patología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Calcificación Vascular/inducido químicamente , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
3.
Acta Pharmacol Sin ; 43(3): 624-633, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34163023

RESUMEN

Vascular calcification (VC) is characterized by pathological depositions of calcium and phosphate in the arteries and veins via an active cell-regulated process, in which vascular smooth muscle cells (VSMCs) transform into osteoblast/chondrocyte-like cells as in bone formation. VC is associated with significant morbidity and mortality in chronic kidney disease (CKD) and cardiovascular disease, but the underlying mechanisms remain unclear. In this study we investigated the role of large-conductance calcium-activated potassium (BK) channels in 3 experimental VC models. VC was induced in vascular smooth muscle cells (VSMCs) by ß-glycerophosphate (ß-GP), or in rats by subtotal nephrectomy, or in mice by high-dosage vitamin D3. We showed that the expression of BK channels in the artery of CKD rats with VC and in ß-GP-treated VSMCs was significantly decreased, which was functionally confirmed by patch-clamp recording. In ß-GP-treated VSMCs, BK channel opener NS1619 (20 µM) significantly alleviated VC by decreasing calcium content and alkaline phosphatase activity. Furthermore, NS1619 decreased mRNA expression of ostoegenic genes OCN and OPN, as well as Runx2 (a key transcription factor involved in preosteoblast to osteoblast differentiation), and increased the expression of α-SMA protein, whereas BK channel inhibitor paxilline (10 µM) caused the opposite effects. In primary cultured VSMCs from BK-/- mice, BK deficiency aggravated calcification as did BK channel inhibitor in normal VSMCs. Moreover, calcification was more severe in thoracic aorta rings of BK-/- mice than in those of wild-type littermates. Administration of BK channel activator BMS191011 (10 mg· kg-1 ·d-1) in high-dosage vitamin D3-treated mice significantly ameliorated calcification. Finally, co-treatment with Akt inhibitor MK2206 (1 µM) or FoxO1 inhibitor AS1842856 (3 µM) in calcified VSMCs abrogated the effects of BK channel opener NS1619. Taken together, activation of BK channels ameliorates VC via Akt/FoxO1 signaling pathways. Strategies to activate BK channels and/or enhance BK channel expression may offer therapeutic avenues to control VC.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Calcificación Vascular/patología , Fosfatasa Alcalina/efectos de los fármacos , Animales , Aorta Torácica/efectos de los fármacos , Bencimidazoles/farmacología , Colecalciferol/farmacología , Modelos Animales de Enfermedad , Glicerofosfatos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Nefrectomía , Osteocalcina/efectos de los fármacos , Osteopontina/efectos de los fármacos , Fragmentos de Péptidos/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
4.
Curr Issues Mol Biol ; 43(2): 1057-1071, 2021 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-34563044

RESUMEN

Postmenopausal women are at an increased risk of vascular calcification which is defined as the pathological deposition of minerals in the vasculature, and is strongly linked with increased cardiovascular disease risk. Since estrogen-replacement therapy is associated with increased cancer risk, there is a strong need for safer therapeutic approaches. In this study we aimed to investigate the protective and therapeutic effects of the phytoestrogen resveratrol against vascular calcification in ovariectomized rats, a preclinical model of postmenopause. Furthermore, we aimed to compare the effects of resveratrol to those of estrogen and to explore the mechanisms underpinning those effects. Treatment with resveratrol or estrogen ameliorated aortic calcification in ovariectomized rats, as shown by reduced calcium deposition in the arterial wall. Mechanistically, the effects of resveratrol and estrogen were mediated via the activation of SIRT1 signaling. SIRT1 protein expression was downregulated in the aortas of ovariectomized rats, and upregulated in rats treated with resveratrol or estrogen. Moreover, resveratrol and estrogen reduced the levels of the osteogenic markers: runt-related transcription factor 2 (RUNX2), osteocalcin and alkaline phosphatase (ALP) which have been shown to play a role during vascular calcification. Additionally, the senescence markers (p53, p16 and p21) which were also reported to play a role in the pathogenesis of vascular calcification, were reduced upon treatment with resveratrol and estrogen. In conclusion, the phytoestrogen resveratrol may be a safer alternative to estrogen, as a therapeutic approach against the progression of vascular calcification during postmenopause.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Fitoestrógenos/farmacología , Resveratrol/farmacología , Transducción de Señal , Sirtuina 1/metabolismo , Calcificación Vascular/tratamiento farmacológico , Animales , Aorta/efectos de los fármacos , Aorta/patología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Femenino , Osteogénesis/efectos de los fármacos , Ovariectomía , Posmenopausia , Ratas , Sirtuina 1/genética , Calcificación Vascular/patología
5.
Aging (Albany NY) ; 13(12): 16749-16762, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34170851

RESUMEN

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.


Asunto(s)
Exosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Fósforo/toxicidad , ARN Largo no Codificante/metabolismo , Calcificación Vascular/genética , Línea Celular , Transdiferenciación Celular/efectos de los fármacos , Análisis por Conglomerados , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Exosomas/ultraestructura , Perfilación de la Expresión Génica , Humanos , Minerales/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Calcificación Vascular/patología
6.
Nutrients ; 13(4)2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33917175

RESUMEN

Osteoporosis (OP) and vascular calcification (VC) represent relevant health problems that frequently coexist in the elderly population. Traditionally, they have been considered independent processes, and mainly age-related. However, an increasing number of studies have reported their possible direct correlation, commonly defined as "bone-vascular crosstalk". Vitamin K2 (VitK2), a family of several natural isoforms also known as menaquinones (MK), has recently received particular attention for its role in maintaining calcium homeostasis. In particular, VitK2 deficiency seems to be responsible of the so-called "calcium paradox" phenomenon, characterized by low calcium deposition in the bone and its accumulation in the vessel wall. Since these events may have important clinical consequences, and the role of VitK2 in bone-vascular crosstalk has only partially been explained, this review focuses on its effects on the bone and vascular system by providing a more recent literature update. Overall, the findings reported here propose the VitK2 family as natural bioactive molecules that could be able to play an important role in the prevention of bone loss and vascular calcification, thus encouraging further in-depth studies to achieve its use as a dietary food supplement.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Resorción Ósea/patología , Huesos/irrigación sanguínea , Calcificación Vascular/patología , Vitamina K 2/farmacología , Animales , Huesos/efectos de los fármacos , Suplementos Dietéticos , Humanos , Vitamina K 2/química
7.
Mol Biol Rep ; 48(1): 887-896, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33394226

RESUMEN

Pathological calcification is a major cause of cardiovascular morbidities primarily in population with chronic kidney disease (CKD), end stage renal diseases (ERSD) and metabolic disorders. Investigators have accepted the fact that vascular calcification is not a passive process but a highly complex, cell mediated, active process in patients with cardiovascular disease (CVD) resulting from, metabolic insults of bone fragility, diabetes, hypertension, dyslipidemia and atherosclerosis. Over the years, studies have revealed various mechanisms of vascular calcification like induction of bone formation, apoptosis, alteration in Ca-P balance and loss of inhibition. Novel clinical studies targeting cellular mechanisms of calcification provide promising and potential avenues for drug development. The interventions include phosphate binders, sodium thiosulphate, vitamin K, calcimimetics, vitamin D, bisphosphonates, Myoinositol hexaphosphate (IP6), Denosumab and TNAP inhibitors. Concurrently investigators are also working towards reversing or curing pathological calcification. This review focuses on the relationship of vascular calcification to clinical diseases, regulators and factors causing calcification including genetics which have been identified. At present, there is lack of any significant preventive measures for calcifications and hence this review explores further possibilities for drug development and treatment modalities.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Diabetes Mellitus/tratamiento farmacológico , Dislipidemias/tratamiento farmacológico , Hipertensión/tratamiento farmacológico , Insuficiencia Renal Crónica/tratamiento farmacológico , Calcificación Vascular/tratamiento farmacológico , Aterosclerosis/metabolismo , Aterosclerosis/patología , Calcimiméticos/uso terapéutico , Calcio/metabolismo , Denosumab/uso terapéutico , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Difosfonatos/uso terapéutico , Dislipidemias/metabolismo , Dislipidemias/patología , Inhibidores Enzimáticos/uso terapéutico , Homeostasis/efectos de los fármacos , Hipertensión/metabolismo , Hipertensión/patología , Fosfatos de Inositol/uso terapéutico , Fósforo/metabolismo , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Tiosulfatos/uso terapéutico , Calcificación Vascular/metabolismo , Calcificación Vascular/patología , Vitamina D/uso terapéutico , Vitamina K/uso terapéutico
8.
Clin Sci (Lond) ; 135(3): 515-534, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33479769

RESUMEN

In chronic kidney disease (CKD), hyperphosphatemia is a key factor promoting medial vascular calcification, a common complication associated with cardiovascular events and high mortality. Vascular calcification involves osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs), but the complex signaling events inducing pro-calcific pathways are incompletely understood. The present study investigated the role of acid sphingomyelinase (ASM)/ceramide as regulator of VSMC calcification. In vitro, both, bacterial sphingomyelinase and phosphate increased ceramide levels in VSMCs. Bacterial sphingomyelinase as well as ceramide supplementation stimulated osteo-/chondrogenic transdifferentiation during control and high phosphate conditions and augmented phosphate-induced calcification of VSMCs. Silencing of serum- and glucocorticoid-inducible kinase 1 (SGK1) blunted the pro-calcific effects of bacterial sphingomyelinase or ceramide. Asm deficiency blunted vascular calcification in a cholecalciferol-overload mouse model and ex vivo isolated-perfused arteries. In addition, Asm deficiency suppressed phosphate-induced osteo-/chondrogenic signaling and calcification of cultured VSMCs. Treatment with the functional ASM inhibitors amitriptyline or fendiline strongly blunted pro-calcific signaling pathways in vitro and in vivo. In conclusion, ASM/ceramide is a critical upstream regulator of vascular calcification, at least partly, through SGK1-dependent signaling. Thus, ASM inhibition by repurposing functional ASM inhibitors to reduce the progression of vascular calcification during CKD warrants further study.


Asunto(s)
Transdiferenciación Celular , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Esfingomielina Fosfodiesterasa/farmacología , Calcificación Vascular/patología , Amitriptilina/farmacología , Animales , Células Cultivadas , Ceramidas/metabolismo , Condrogénesis/efectos de los fármacos , Fendilina/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Fosfatos/farmacología
9.
Cell Death Dis ; 11(11): 991, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33203874

RESUMEN

Pyruvate dehydrogenase kinase 4 (PDK4) is an important mitochondrial matrix enzyme in cellular energy regulation. Previous studies suggested that PDK4 is increased in the calcified vessels of patients with atherosclerosis and is closely associated with mitochondrial function, but the precise regulatory mechanisms remain largely unknown. This study aims to investigate the role of PDK4 in vascular calcification and the molecular mechanisms involved. Using a variety of complementary techniques, we found impaired autophagic activity in the process of vascular smooth muscle cells (VSMCs) calcification, whereas knocking down PDK4 had the opposite effect. PDK4 drives the metabolic reprogramming of VSMCs towards a Warburg effect, and the inhibition of PDK4 abrogates VSMCs calcification. Mechanistically, PDK4 disturbs the integrity of the mitochondria-associated endoplasmic reticulum membrane, concomitantly impairing mitochondrial respiratory capacity, which contributes to a decrease in lysosomal degradation by inhibiting the V-ATPase and lactate dehydrogenase B interaction. PDK4 also inhibits the nuclear translocation of the transcription factor EB, thus inhibiting lysosomal function. These changes result in the interruption of autophagic flux, which accelerates calcium deposition in VSMCs. In addition, glycolysis serves as a metabolic adaptation to improve VSMCs oxidative stress resistance, whereas inhibition of glycolysis by 2-deoxy-D-glucose induces the apoptosis of VSMCs and increases the calcium deposition in VSMCs. Our results suggest that PDK4 plays a key role in vascular calcification through autophagy inhibition and metabolic reprogramming.


Asunto(s)
Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Calcificación Vascular/metabolismo , Animales , Autofagia/fisiología , Señalización del Calcio , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Calcificación Vascular/patología
10.
Medicine (Baltimore) ; 99(42): e22813, 2020 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-33080758

RESUMEN

RATIONALE: Idiopathic mesenteric phlebosclerosis (IMP) is a rare form of ischemic colitis. It is more common in the Asian population people with Asian ancestry. Disease pathogenesis and etiology are not fully elucidated but may be associated with the long-term intake of toxins and other substances, including Chinese herbs. The disease has typical radiological and endoscopic features. Radiologic examination combined with endoscopy can lead to a conclusive diagnosis. PATIENT CONCERNS: We present 2 cases of IMP: in male patients aged 66 and 79 years. The first patient presented with diarrhea and abdominal pain, and the second patient presented with numbness of limbs and abdominal discomfort. These patients had a history of long-term use of Chinese herbal medicine (CHM). DIAGNOSIS: Both patients were diagnosed with IMP by endoscopy and radiology, and the diagnosis confirmed by biopsy in the first patient. INTERVENTIONS: The first patient was advised to stop using CHM. Both patients were given conservative treatment and were followed up regularly. OUTCOMES: Symptoms improved after conservative treatment. The patients had no obvious discomfort during the follow-up period. CONCLUSION: We suspect that the disease is induced by the long-term use of CHM, and dosage and duration of use may determine disease severity.


Asunto(s)
Colitis Isquémica/inducido químicamente , Medicamentos Herbarios Chinos/efectos adversos , Venas Mesentéricas/diagnóstico por imagen , Venas Mesentéricas/patología , Calcificación Vascular/inducido químicamente , Anciano , Colitis Isquémica/diagnóstico por imagen , Colitis Isquémica/patología , Humanos , Mucosa Intestinal/patología , Masculino , Esclerosis , Tomografía Computarizada por Rayos X , Calcificación Vascular/diagnóstico por imagen , Calcificación Vascular/patología
11.
Int J Mol Sci ; 21(12)2020 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-32630604

RESUMEN

Vascular calcification describes the formation of mineralized tissue within the blood vessel wall, and it is highly associated with increased cardiovascular morbidity and mortality in patients with chronic kidney disease, diabetes, and atherosclerosis. In this article, we briefly review different rodent models used to study vascular calcification in vivo, and critically assess the strengths and weaknesses of the current techniques used to analyze and quantify calcification in these models, namely 2-D histology and the o-cresolphthalein assay. In light of this, we examine X-ray micro-computed tomography (µCT) as an emerging complementary tool for the analysis of vascular calcification in animal models. We demonstrate that this non-destructive technique allows us to simultaneously quantify and localize calcification in an intact vessel in 3-D, and we consider recent advances in µCT sample preparation techniques. This review also discusses the potential to combine 3-D µCT analyses with subsequent 2-D histological, immunohistochemical, and proteomic approaches in correlative microscopy workflows to obtain rich, multifaceted information on calcification volume, calcification load, and signaling mechanisms from within the same arterial segment. In conclusion we briefly discuss the potential use of µCT to visualize and measure vascular calcification in vivo in real-time.


Asunto(s)
Calcificación Vascular/patología , Microtomografía por Rayos X/métodos , Microtomografía por Rayos X/tendencias , Animales , Aterosclerosis/patología , Humanos , Imagenología Tridimensional/métodos , Microscopía/métodos , Modelos Animales , Proteómica , Insuficiencia Renal Crónica/patología , Calcificación Vascular/diagnóstico por imagen , Calcificación Vascular/metabolismo
12.
Circ Genom Precis Med ; 13(3): e002710, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32397743

RESUMEN

BACKGROUND: Aortic stenosis (AS) contributes to cardiovascular mortality and morbidity but disease mechanisms remain largely unknown. Recent evidence associates a single nucleotide polymorphism rs174547 within the FADS1 gene, encoding FADS1 (fatty acid desaturase 1), with risk of several cardiovascular outcomes, including AS. FADS1 encodes a rate-limiting enzyme for ω-3 and ω-6 fatty acid metabolism. The aim of this study was to decipher the local transcriptomic and lipidomic consequences of rs174547 in tricuspid aortic valves from patients with AS. METHODS: Expression quantitative trait loci study was performed using data from Illumina Human610-Quad BeadChip, Infinium Global Screening Arrays, and Affymetrix Human Transcriptome 2.0 arrays in calcified and noncalcified aortic valve tissue from 58 patients with AS (mean age, 74.2; SD, 5.9). Fatty acid content was assessed in aortic valves from 25 patients with AS using gas chromatography. Δ5 and Δ6 desaturase activity was assessed by the product-to-precursor ratio. RESULTS: The minor C-allele of rs174547, corresponding to the protective genotype for AS, was associated with higher FADS2 mRNA levels in calcified valve tissue, whereas FADS1 mRNA and other transcripts in proximity of the single nucleotide polymorphism were unaltered. In contrast, the FADS1 Δ5-desaturase activity and the FADS2 Δ6-desaturase activity were decreased. Finally, docosahexaenoic acid was decreased in calcified tissue compared with non-calcified tissue and C-allele carriers exhibited increased docosahexaenoic acid levels. Overall desaturase activity measured with ω-3 fatty acids was higher in C-allele carriers. CONCLUSIONS: The association between the FADS1 genotype and AS may implicate effects on valvular fatty acids.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Calcinosis/metabolismo , Ácido Graso Desaturasas/biosíntesis , Ácidos Grasos Omega-3/metabolismo , Ácidos Grasos Omega-6/metabolismo , Regulación Enzimológica de la Expresión Génica , Calcificación Vascular/metabolismo , Anciano , Anciano de 80 o más Años , Estenosis de la Válvula Aórtica/patología , Calcinosis/patología , delta-5 Desaturasa de Ácido Graso , Femenino , Humanos , Masculino , Calcificación Vascular/patología
13.
Int J Mol Sci ; 21(7)2020 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-32252330

RESUMEN

Vascular calcification, which involves the deposition of calcifying particles within the arterial wall, is mediated by atherosclerosis, vascular smooth muscle cell osteoblastic changes, adventitial mesenchymal stem cell osteoblastic differentiation, and insufficiency of the calcification inhibitors. Recent observations implied a role for mesenchymal stem cells and endothelial progenitor cells in vascular calcification. Mesenchymal stem cells reside in the bone marrow and the adventitial layer of arteries. Endothelial progenitor cells that originate from the bone marrow are an important mechanism for repairing injured endothelial cells. Mesenchymal stem cells may differentiate osteogenically by inflammation or by specific stimuli, which can activate calcification. However, the bioactive substances secreted from mesenchymal stem cells have been shown to mitigate vascular calcification by suppressing inflammation, bone morphogenetic protein 2, and the Wingless-INT signal. Vitamin D deficiency may contribute to vascular calcification. Vitamin D supplement has been used to modulate the osteoblastic differentiation of mesenchymal stem cells and to lessen vascular injury by stimulating adhesion and migration of endothelial progenitor cells. This narrative review clarifies the role of mesenchymal stem cells and the possible role of vitamin D in the mechanisms of vascular calcification.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Calcificación Vascular/etiología , Calcificación Vascular/metabolismo , Vitamina D/metabolismo , Animales , Biomarcadores , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Células Progenitoras Endoteliales/efectos de los fármacos , Humanos , Inmunofenotipificación , Células Madre Mesenquimatosas/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Pericitos/efectos de los fármacos , Pericitos/metabolismo , Calcificación Vascular/tratamiento farmacológico , Calcificación Vascular/patología , Vitamina D/farmacología , Vitamina D/uso terapéutico
14.
Nutrients ; 12(3)2020 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-32213826

RESUMEN

Abdominal aortic calcification (AAC) detected on lateral vertebral fracture assessment is associated with increased cardiovascular risk. Vitamin D deficiency and toxicity have been linked with vascular calcification. The objective of this study was to determine the effect of high-dose vitamin D on the progression of AAC. The Physical Performance, Osteoporosis and vitamin D in African American Women (PODA) is a randomized, clinical trial examining the effect of vitamin D. There were 14.7% subjects with AAC in the vitamin D group, compared to 12.1% in the placebo group at baseline. The prevalence of extended AAC at baseline was 6.4% in the vitamin D group and 3.5% in the placebo group. The extended calcification scores over time were not different between groups. There was no association between AAC and serum 25(OH)D. However, PTH was associated with an increase in AAC in the placebo group.


Asunto(s)
Aorta Abdominal , Negro o Afroamericano , Calcificación Vascular/epidemiología , Calcificación Vascular/etiología , Vitamina D/administración & dosificación , Anciano , Aorta Abdominal/patología , Biomarcadores , Suplementos Dietéticos , Susceptibilidad a Enfermedades , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Osteoporosis , Prevalencia , Factores de Riesgo , Factores Sexuales , Calcificación Vascular/metabolismo , Calcificación Vascular/patología , Vitamina D/sangre
16.
Eur J Pharm Biopharm ; 150: 108-119, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32151731

RESUMEN

Generalized arterial calcification of infancy (GACI) and pseudoxanthoma elasticum (PXE) are characterized by pathologic calcifications in the media of large- and medium sized arteries. GACI is associated with biallelic mutations in ENPP1 in the majority of cases, whereas mutations in ABCC6 are known to cause PXE. Different treatment approaches including bisphosphonates and orally administered pyrophosphate (PPi) were investigated in recent years, but reversion of calcification could not be achieved. With this study, we pursued the idea of a combination of controlled drug delivery through nanoparticles and active targeting via antibody conjugation to develop a treatment for GACI and PXE. To establish a suitable drug delivery system, the chelating drug diethylenetriamine pentaacetic acid (DTPA) was conjugated to nanoparticles composed of human serum albumin (HSA) as biodegradable and non-toxic particle matrix. To accomplish an active targeting of the elastic fibers exposed through calcification of the affected areas, the nanoparticle surface was functionalized with an anti-elastin antibody. Cytotoxicity and cell interaction studies revealed favorable preconditions for the intended i.v. application. The chelating ability was evaluated in vitro and ex vivo on aortic ring culture isolated from two mouse models of GACI and PXE. The positive results led to the conclusion that the produced nanoparticles might be a promising therapy in the treatment of GACI and PXE.


Asunto(s)
Anticuerpos/química , Aorta/efectos de los fármacos , Quelantes del Calcio/farmacología , Portadores de Fármacos , Elastina/inmunología , Ácido Pentético/farmacología , Seudoxantoma Elástico/tratamiento farmacológico , Albúmina Sérica Humana/química , Calcificación Vascular/tratamiento farmacológico , Animales , Anticuerpos/inmunología , Aorta/inmunología , Aorta/metabolismo , Aorta/patología , Quelantes del Calcio/química , Línea Celular , Composición de Medicamentos , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/deficiencia , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Nanopartículas , Ácido Pentético/química , Seudoxantoma Elástico/inmunología , Seudoxantoma Elástico/metabolismo , Seudoxantoma Elástico/patología , Albúmina Sérica Humana/metabolismo , Calcificación Vascular/inmunología , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
17.
Nephrol Dial Transplant ; 35(Suppl 2): ii31-ii37, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32162665

RESUMEN

Chronic kidney disease (CKD) is a clinical model of premature ageing characterized by cardiovascular disease, persistent uraemic inflammation, osteoporosis muscle wasting and frailty. The accelerated early vascular ageing (EVA) process mediated by medial vascular calcification (VC) is a hallmark of senescence as well as a strong predictor of cardiovascular morbidity and mortality in the CKD population. Current clinical therapeutic strategies and novel treatments for VC have not yet been proven to prevent or reverse VC progression in patients with CKD. Knowledge of the fundamental mechanism underlying EVA is urgently needed to identify and develop novel and efficient therapeutic targets for VC and EVA. An accumulating body of evidence indicates that deoxyribonucleic acid (DNA) damage-induced cellular senescence and 'inflammaging' may largely contribute to such pathological conditions characterized by accelerated EVA. Growing evidence shows that nuclear factor erythroid 2-related factor 2 (NRF2) signalling and vitamin K play a crucial role in counteracting oxidative stress, DNA damage, senescence and inflammaging, whereby NRF2 activation and vitamin K supplementation may provide a novel treatment target for EVA. In this review we discuss the link between senescence and EVA in the context of CKD, with a focus on the role of NRF2 and vitamin K in DNA damage signalling, senescence and inflammaging.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Senescencia Celular , Daño del ADN , Inflamación/fisiopatología , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/etiología , Vitamina K/metabolismo , Enfermedades Cardiovasculares/patología , Progresión de la Enfermedad , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Calcificación Vascular/patología
18.
Sci Rep ; 10(1): 1645, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32015399

RESUMEN

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.


Asunto(s)
Ceramidasa Ácida/metabolismo , Calcificación Vascular/metabolismo , Ceramidasa Ácida/genética , Animales , Aorta/metabolismo , Aorta/patología , Señalización del Calcio , Células Cultivadas , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patología , Lipogranulomatosis de Farber/genética , Lipogranulomatosis de Farber/metabolismo , Lisosomas/metabolismo , Masculino , Redes y Vías Metabólicas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Cardiovasculares , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Esfingolípidos/metabolismo , Canales de Potencial de Receptor Transitorio/agonistas , Canales de Potencial de Receptor Transitorio/metabolismo , Calcificación Vascular/genética , Calcificación Vascular/patología
19.
Biomed Phys Eng Express ; 6(5): 055011, 2020 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-33444242

RESUMEN

Computed tomography (CT) is the reference method for cardiac imaging, but concerns have been raised regarding the radiation dose of CT examinations. Recently, photon counting detectors (PCDs) and interior tomography, in which the radiation beam is limited to the organ-of-interest, have been suggested for patient dose reduction. In this study, we investigated interior PCD-CT (iPCD-CT) for non-enhanced quantification of coronary artery calcium (CAC) using an anthropomorphic torso phantom and ex vivo coronary artery samples. We reconstructed the iPCD-CT measurements with filtered back projection (FBP), iterative total variation (TV) regularization, padded FBP, and adaptively detruncated FBP and adaptively detruncated TV. We compared the organ doses between conventional CT and iPCD-CT geometries, assessed the truncation and cupping artifacts with iPCD-CT, and evaluated the CAC quantification performance of iPCD-CT. With approximately the same effective dose between conventional CT geometry (0.30 mSv) and interior PCD-CT with 10.2 cm field-of-view (0.27 mSv), the organ dose of the heart was increased by 52.3% with interior PCD-CT when compared to CT. Conversely, the organ doses to peripheral and radiosensitive organs, such as the stomach (55.0% reduction), were often reduced with interior PCD-CT. FBP and TV did not sufficiently reduce the truncation artifact, whereas padded FBP and adaptively detruncated FBP and TV yielded satisfactory truncation artifact reduction. Notably, the adaptive detruncation algorithm reduced truncation artifacts effectively when it was combined with reconstruction detrending. With this approach, the CAC quantification accuracy was good, and the coronary artery disease grade reclassification rate was particularly low (5.6%). Thus, our results confirm that CAC quantification can be performed with the interior CT geometry, that the artifacts are effectively reduced with suitable interior reconstruction methods, and that interior tomography provides efficient patient dose reduction.


Asunto(s)
Calcio/metabolismo , Enfermedad de la Arteria Coronaria/fisiopatología , Procesamiento de Imagen Asistido por Computador/métodos , Fantasmas de Imagen , Fotones , Tomografía Computarizada por Rayos X/métodos , Calcificación Vascular/patología , Adulto , Algoritmos , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Corazón/diagnóstico por imagen , Corazón/efectos de la radiación , Humanos , Masculino , Dosis de Radiación , Calcificación Vascular/diagnóstico por imagen , Calcificación Vascular/metabolismo
20.
Nefrologia (Engl Ed) ; 40(3): 336-344, 2020.
Artículo en Inglés, Español | MEDLINE | ID: mdl-31864861

RESUMEN

BACKGROUND: The impact of hepatitis virus infection on arterial calcification (AC) was not studied. OBJECTIVE: To study the prevalence, severity and distribution of AC in incident hemodialysis patients with hepatitis B and C viral infection. CASES AND METHODS: 172 stage 5 CKD adults (98 male and 74 female) were included; 58 of them were seronegative for both hepatitis B and C (SN group), 48 were positive for hepatitis B virus infection (HBV group) and 66 were hepatitis C virus positive (HCV group). Beside histopathology of the obtained arterial samples, all these cases were examined for body mass index (BMI), serum calcium (Ca), phosphorus (P), alkaline phosphatase (AP), serum albumin, uric acid (UA), alanine transaminase (ALT), parathormone (PTH), fibroblast growth factor 23(FGF23), interleukin 6 (IL6), and 25 hydroxy vitamin D (25 (OH) vit D), hemoglobin concentration, and serum ferritin. RESULTS: 86 (50%) of the cases had AC; 11 of them were in SN group (19%), 9 in HBV group (18.8%) and all the 66 HCV group (100%). In SN group, 4 had intimal calcification, 5 had medial calcification, and 2 had both intimal and medial calcification. In HBV group, 9 had intimal calcification, while no cases were encountered with either medial or both site calcifications. In HCV group, 16 had intimal calcification, 31 had medial calcification, and 19 had both intimal and medial calcification. Calcification was in the form of spots in one case in SN group, and 6 cases in HBV group, a single plaque of calcification in 5 cases of SN group, 3 cases of HBV group, and 16 cases of HCV group, multiple plaques were detected in 4 cases in SN group, and 31 cases in HCV group, and diffuse calcification in one case in SN group, and 19 cases in HCV group. In HBV group, calcification was only detected in patients with high viremia, while all patients with low or moderate viremia were devoid of calcification. In HCV group, all patients with low viremia had intimal solitary plaque of calcification, all patients with moderate viremia had multiple plaques of medial calcification, while all patients with high viremia had diffuse intimal and medial calcification. Both groups of viral hepatitis were significantly different in comparison to SN group in either distribution or calcification score (P<0.001 in all). HBV group had significantly lower serum P, CaxP and PTH in comparison to SN group (4.6±0.66 vs. 5.45±0.77mg/dL, 36.4±7.2 vs. 44.1±8.69, and 348±65.4 vs. 405.9±83.2pg/mL, P<0.001, <0.001, and 0.035 respectively). On the other hand, HCV group did not show any significant difference in any of the studied parameters compared to SN group. CONCLUSION: HCV positive patients are more prone to develop AC that is more extensive. HBV positive patients were less likely to have arterial medial calcification, probably related to lower serum phosphorus, CaxP product and PTH. HCV infection should be added as risk factor for AC among CKD patients. Further studies are needed to confirm these findings.


Asunto(s)
Arteriopatías Oclusivas/epidemiología , Hepatitis B/complicaciones , Hepatitis C/complicaciones , Diálisis Renal , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/epidemiología , Viremia/complicaciones , Adulto , Arteriopatías Oclusivas/sangre , Arteriopatías Oclusivas/etiología , Arteriopatías Oclusivas/patología , Proteínas Sanguíneas/análisis , Calcio/análisis , Susceptibilidad a Enfermedades , Femenino , Factor-23 de Crecimiento de Fibroblastos , Hepatitis B/sangre , Hepatitis C/sangre , Humanos , Masculino , Persona de Mediana Edad , Hormona Paratiroidea/sangre , Fósforo/sangre , Prevalencia , Arteria Radial/química , Arteria Radial/patología , Insuficiencia Renal Crónica/sangre , Factores de Riesgo , Índice de Severidad de la Enfermedad , Túnica Íntima/química , Túnica Media/química , Calcificación Vascular/sangre , Calcificación Vascular/etiología , Calcificación Vascular/patología , Viremia/sangre , Vitamina D/sangre , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA