Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Methods Mol Biol ; 2664: 317-331, 2023.
Article En | MEDLINE | ID: mdl-37423997

Vascular calcification is an active pathological process, characterised by cellular dysregulation and subsequent changes to the extracellular environment. In vivo detection of vascular calcification is only possible late stage via computed tomography, and there is no single biomarker for detecting progression of vascular calcification. There is an unmet clinical need to determine progression of vascular calcification in vulnerable patients. This is especially needed in chronic kidney disease (CKD) patients where there is a correlation of cardiovascular disease with declining renal status. We hypothesised that the entirety of circulating components should be taken into consideration with vessel wall cells to determine real-time vascular calcification development. In this protocol we describe the isolation and characterisation of human primary vascular smooth muscle cells (hpVSMCs), and the addition of human serum or plasma to hpVSMCs in a calcification assay and analysis. The BioHybrid analysis of biological changes to in vitro hpVSMC calcification is reflective of in vivo vascular calcification status. We suggest this analysis can discriminate between CKD patient cohorts and has the potential for wider application for risk factor determination in CKD and the general population.


Kidney Failure, Chronic , Renal Insufficiency, Chronic , Vascular Calcification , Humans , Kidney/pathology , Renal Insufficiency, Chronic/complications , Calcification, Physiologic , Myocytes, Smooth Muscle/pathology
2.
Int J Mol Sci ; 23(24)2022 Dec 17.
Article En | MEDLINE | ID: mdl-36555778

Cardiovascular disease is the major cause of death worldwide. The success of medication and other preventive measures introduced in the last century have not yet halted the epidemic of cardiovascular disease. Although the molecular mechanisms of the pathophysiology of the heart and vessels have been extensively studied, the burden of ischemic cardiovascular conditions has risen to become a top cause of morbidity and mortality. Calcium has important functions in the cardiovascular system. Calcium is involved in the mechanism of excitation-contraction coupling that regulates numerous events, ranging from the production of action potentials to the contraction of cardiomyocytes and vascular smooth muscle cells. Both in the heart and vessels, the rise of intracellular calcium is sensed by calmodulin, a protein that regulates and activates downstream kinases involved in regulating calcium signalling. Among them is the calcium calmodulin kinase family, which is involved in the regulation of cardiac functions. In this review, we present the current literature regarding the role of calcium/calmodulin pathways in the heart and vessels with the aim to summarize our mechanistic understanding of this process and to open novel avenues for research.


Calmodulin , Cardiovascular Diseases , Humans , Calmodulin/metabolism , Calcium/metabolism , Cardiovascular Diseases/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
3.
J Vis Exp ; (184)2022 06 02.
Article En | MEDLINE | ID: mdl-35723489

Vascular calcification involves a series of degenerative pathologies, including inflammation, changes to cellular phenotype, cell death, and the absence of calcification inhibitors, that concomitantly lead to a loss of vessel elasticity and function. Vascular calcification is an important contributor to morbidity and mortality in many pathologies, including chronic kidney disease, diabetes mellitus, and atherosclerosis. Current research models to study vascular calcification are limited and are only viable at the late stages of calcification development in vivo. In vitro tools for studying vascular calcification use end-point measurements, increasing the demands on biological material and risking the introduction of variability to research studies. We demonstrate the application of a novel fluorescently labeled probe that binds to in vitro calcification development on human vascular smooth muscle cells and determines the real-time development of in vitro calcification. In this protocol, we describe the application of our newly developed calcification assay, a novel tool in disease modeling that has potential translational applications. We envisage this assay to be relevant in a broader spectrum of mineral deposition research, including applications in bone, cartilage, or dental research.


Renal Insufficiency, Chronic , Vascular Calcification , Calcium/metabolism , Humans , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology
4.
Kidney Int ; 101(2): 338-348, 2022 02.
Article En | MEDLINE | ID: mdl-34774554

Chronic kidney disease (CKD) is accompanied with extensive cardiovascular calcification, in part correlating with functional vitamin K deficiency. Here, we sought to determine causes for vitamin K deficiency beyond reduced dietary intake. Initially, vitamin K uptake and distribution into circulating lipoproteins after a single administration of vitamin K1 plus K2 (menaquinone 4 and menaquinone 7, respectively) was determined in patients on dialysis therapy and healthy individuals. The patients incorporated very little menaquinone 7 but more menaquinone 4 into high density lipoprotein (HDL) and low-density lipoprotein particles than did healthy individuals. In contrast to healthy persons, HDL particles from the patients could not be spiked with menaquinone 7 in vitro and HDL uptake was diminished in osteoblasts. A reduced carboxylation activity (low vitamin K activity) of uremic HDL particles spiked with menaquinone 7 vs. that of controls was confirmed in a bioassay using human primary vascular smooth muscle cells. Kidney menaquinone 4 tissue levels were reduced in 5/6-nephrectomized versus sham-operated C57BL/6 mice after four weeks of a vitamin K rich diet. From the analyzed enzymes involved in vitamin K metabolism, kidney HMG-CoA reductase protein was reduced in both rats and patients with CKD. In a trial on the efficacy and safety of atorvastatin in 1051 patients with type 2 diabetes receiving dialysis therapy, no pronounced vitamin K deficiency was noted. However, the highest levels of PIVKA-II (biomarker of subclinical vitamin K deficiency) were noted when a statin was combined with a proton pump inhibitor. Thus, profound disturbances in lipoprotein mediated vitamin K transport and metabolism in uremia suggest that menaquinone 7 supplementation to patients on dialysis therapy has reduced efficacy.


Renal Insufficiency, Chronic , Vitamin K Deficiency , Vitamin K/metabolism , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Humans , Mice , Mice, Inbred C57BL , Rats , Renal Insufficiency, Chronic/metabolism , Tissue Distribution , Vitamin K/therapeutic use , Vitamin K 1/metabolism , Vitamin K 1/therapeutic use , Vitamin K 2/metabolism , Vitamin K 2/therapeutic use , Vitamin K Deficiency/complications , Vitamin K Deficiency/metabolism
5.
Nutrients ; 13(10)2021 Oct 01.
Article En | MEDLINE | ID: mdl-34684491

Vitamin K dependent proteins (VKDP), such as hepatic coagulation factors and vascular matrix Gla protein (MGP), play key roles in maintaining physiological functions. Vitamin K deficiency results in inactive VKDP and is strongly linked to vascular calcification (VC), one of the major risk factors for cardiovascular morbidity and mortality. In this study we investigated how two vitamin K surrogate markers, dephosphorylated-undercarboxylated MGP (dp-ucMGP) and protein induced by vitamin K absence II (PIVKA-II), reflect vitamin K status in patients on hemodialysis or with calcific uremic arteriolopathy (CUA) and patients with atrial fibrillation or aortic valve stenosis. Through inter- and intra-cohort comparisons, we assessed the influence of vitamin K antagonist (VKA) use, vitamin K supplementation and disease etiology on vitamin K status, as well as the correlation between both markers. Overall, VKA therapy was associated with 8.5-fold higher PIVKA-II (0.25 to 2.03 AU/mL) and 3-fold higher dp-ucMGP (843 to 2642 pM) levels. In the absence of VKA use, non-renal patients with established VC have dp-ucMGP levels similar to controls (460 vs. 380 pM), while in HD and CUA patients, levels were strongly elevated (977 pM). Vitamin K supplementation significantly reduced dp-ucMGP levels within 12 months (440 to 221 pM). Overall, PIVKA-II and dp-ucMGP showed only weak correlation (r2 ≤ 0.26) and distinct distribution pattern in renal and non-renal patients. In conclusion, VKA use exacerbated vitamin K deficiency across all etiologies, while vitamin K supplementation resulted in a vascular VKDP status better than that of the general population. Weak correlation of vitamin K biomarkers calls for thoughtful selection lead by the research question. Vitamin K status in non-renal deficient patients was not anomalous and may question the role of vitamin K deficiency in the pathogenesis of VC in these patients.


Biomarkers/blood , Calcium-Binding Proteins/blood , Extracellular Matrix Proteins/blood , Protein Precursors/blood , Vascular Calcification/blood , Vitamin K Deficiency/blood , Vitamin K/blood , 4-Hydroxycoumarins/therapeutic use , Aortic Valve Stenosis/blood , Aortic Valve Stenosis/complications , Atrial Fibrillation/blood , Atrial Fibrillation/complications , Cardiovascular Diseases/etiology , Cohort Studies , Dietary Supplements , Female , Heart Disease Risk Factors , Humans , Indenes/therapeutic use , Liver/metabolism , Male , Middle Aged , Nutritional Status , Prothrombin , Renal Dialysis/adverse effects , Uremia/blood , Uremia/complications , Vascular Calcification/complications , Vitamin K/antagonists & inhibitors , Vitamin K/therapeutic use , Vitamin K Deficiency/complications , Matrix Gla Protein
6.
Toxins (Basel) ; 12(10)2020 09 29.
Article En | MEDLINE | ID: mdl-33003628

The cardiorenal syndrome relates to the detrimental interplay between the vascular system and the kidney. The uremic milieu induced by reduced kidney function alters the phenotype of vascular smooth muscle cells (VSMC) and promotes vascular calcification, a condition which is strongly linked to cardiovascular morbidity and mortality. Biological mechanisms involved include generation of reactive oxygen species, inflammation and accelerated senescence. A better understanding of the vasotoxic effects of uremic retention molecules may reveal novel avenues to reduce vascular calcification in CKD. The present review aims to present a state of the art on the role of uremic toxins in pathogenesis of vascular calcification. Evidence, so far, is fragmentary and limited with only a few uremic toxins being investigated, often by a single group of investigators. Experimental heterogeneity furthermore hampers comparison. There is a clear need for a concerted action harmonizing and standardizing experimental protocols and combining efforts of basic and clinical researchers to solve the complex puzzle of uremic vascular calcification.


Cardio-Renal Syndrome/metabolism , Kidney/metabolism , Muscle, Smooth, Vascular/metabolism , Renal Insufficiency, Chronic/metabolism , Toxins, Biological/metabolism , Uremia/metabolism , Vascular Calcification/metabolism , Animals , Cardio-Renal Syndrome/pathology , Cardio-Renal Syndrome/physiopathology , Cardio-Renal Syndrome/therapy , Humans , Kidney/pathology , Kidney/physiopathology , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Prognosis , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/therapy , Uremia/pathology , Uremia/physiopathology , Uremia/therapy , Vascular Calcification/pathology , Vascular Calcification/physiopathology , Vascular Calcification/therapy
7.
Front Cell Dev Biol ; 8: 618760, 2020.
Article En | MEDLINE | ID: mdl-33585456

Development of clinical stem cell interventions are hampered by immature cell progeny under current protocols. Human mesenchymal stem cells (hMSCs) are characterized by their ability to self-renew and differentiate into multiple lineages. Generating hMSCs from pluripotent stem cells (iPSCs) is an attractive avenue for cost-efficient and scalable production of cellular material. In this study we generate mature osteoblasts from iPSCs using a stable expandable MSC intermediate, refining established protocols. We investigated the timeframe and phenotype of cells under osteogenic conditions as well as the effect of menaquinone-7 (MK-7) on differentiation. From day 2 we noted a significant increase in RUNX2 expression under osteogenic conditions with MK-7, as well as decreases in ROS species production, increased cellular migration and changes to dynamics of collagen deposition when compared to differentiated cells that were not treated with MK-7. At day 21 OsteoMK-7 increased alkaline phosphatase activity and collagen deposition, as well as downregulated RUNX2 expression, suggesting to a mature cellular phenotype. Throughout we note no changes to expression of osteocalcin suggesting a non-canonical function of MK-7 in osteoblast differentiation. Together our data provide further mechanistic insight between basic and clinical studies on extrahepatic activity of MK-7. Our findings show that MK-7 promotes osteoblast maturation thereby increasing osteogenic differentiation.

...