Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Nutrients ; 15(6)2023 Mar 18.
Article in English | MEDLINE | ID: mdl-36986200

ABSTRACT

This study was designed to investigate the controversy on the potential role of sKlotho as an early biomarker in Chronic Kidney Disease-Mineral Bone Disorder (CKD-MBD), to assess whether sKlotho is a reliable marker of kidney α-Klotho, to deepen the effects of sKlotho on vascular smooth muscle cells (VSMCs) osteogenic differentiation and to evaluate the role of autophagy in this process. Experimental studies were conducted in CKD mice fed a normal phosphorus (CKD+NP) or high phosphorus (CKD+HP) diet for 14 weeks. The patients' study was performed in CKD stages 2-5 and in vitro studies which used VSMCs exposed to non-calcifying medium or calcifying medium with or without sKlotho. The CKD experimental model showed that the CKD+HP group reached the highest serum PTH, P and FGF23 levels, but the lowest serum and urinary sKlotho levels. In addition, a positive correlation between serum sKlotho and kidney α-Klotho was found. CKD mice showed aortic osteogenic differentiation, together with increased autophagy. The human CKD study showed that the decline in serum sKlotho is previous to the rise in FGF23. In addition, both serum sKlotho and FGF23 levels correlated with kidney function. Finally, in VSMCs, the addition of sKlotho prevented osteogenic differentiation and induced autophagy. It can be concluded that serum sKlotho was the earliest CKD-MBD biomarker, a reliable indicator of kidney α-Klotho and that might protect against osteogenic differentiation by increasing autophagy. Nevertheless, further studies are needed to investigate the mechanisms of this possible protective effect.


Subject(s)
Chronic Kidney Disease-Mineral and Bone Disorder , Renal Insufficiency, Chronic , Humans , Mice , Animals , Klotho Proteins , Glucuronidase , Osteogenesis , Fibroblast Growth Factors , Kidney , Phosphorus , Minerals , Biomarkers
2.
Int J Mol Sci ; 24(6)2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36982322

ABSTRACT

Fibrosis plays an important role in the pathogenesis of long-term diabetic complications and contributes to the development of cardiac and renal dysfunction. The aim of this experimental study, performed in a long-term rat model, which resembles type 1 diabetes mellitus, was to investigate the role of soluble Klotho (sKlotho), advanced glycation end products (AGEs)/receptor for AGEs (RAGE), fibrotic Wnt/ß-catenin pathway, and pro-fibrotic pathways in kidney and heart. Diabetes was induced by streptozotocin. Glycaemia was maintained by insulin administration for 24 weeks. Serum and urine sKlotho, AGEs, soluble RAGE (sRAGE) and biochemical markers were studied. The levels of Klotho, RAGEs, ADAM10, markers of fibrosis (collagen deposition, fibronectin, TGF-ß1, and Wnt/ß-catenin pathway), hypertrophy of the kidney and/or heart were analysed. At the end of study, diabetic rats showed higher levels of urinary sKlotho, AGEs and sRAGE and lower serum sKlotho compared with controls without differences in the renal Klotho expression. A significant positive correlation was found between urinary sKlotho and AGEs and urinary albumin/creatinine ratio (uACR). Fibrosis and RAGE levels were significantly higher in the heart without differences in the kidney of diabetic rats compared to controls. The results also suggest the increase in sKlotho and sRAGE excretion may be due to polyuria in the diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental , Kidney Diseases , Rats , Animals , beta Catenin , Receptor for Advanced Glycation End Products , Fibrosis , Glycation End Products, Advanced
3.
Nephrol Dial Transplant ; 38(7): 1729-1740, 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-36722155

ABSTRACT

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.


Subject(s)
MicroRNAs , Vascular Calcification , Animals , Humans , Rats , Biomarkers , Calcium , MicroRNAs/genetics , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Osteogenesis/genetics , Phosphorus , Vascular Calcification/genetics
4.
Nutrients ; 14(13)2022 Jun 22.
Article in English | MEDLINE | ID: mdl-35807767

ABSTRACT

In chronic kidney disease, systemic inflammation and high serum phosphate (P) promote the de-differentiation of vascular smooth muscle cells (VSMC) to osteoblast-like cells, increasing the propensity for medial calcification and cardiovascular mortality. Vascular microRNA-145 (miR-145) content is essential to maintain VSMC contractile phenotype. Because vitamin D induces aortic miR-145, uremia and high serum P reduce it and miR-145 directly targets osteogenic osterix in osteoblasts, this study evaluated a potential causal link between vascular miR-145 reductions and osterix-driven osteogenic differentiation and its counter-regulation by vitamin D. Studies in aortic rings from normal rats and in the rat aortic VSMC line A7r5 exposed to calcifying conditions corroborated that miR-145 reductions were associated with decreases in contractile markers and increases in osteogenic differentiation and calcium (Ca) deposition. Furthermore, miR-145 silencing enhanced Ca deposition in A7r5 cells exposed to calcifying conditions, while miR-145 overexpression attenuated it, partly through increasing α-actin levels and reducing osterix-driven osteogenic differentiation. In mice, 14 weeks after the induction of renal mass reduction, both aortic miR-145 and α-actin mRNA decreased by 80% without significant elevations in osterix or Ca deposition. Vitamin D treatment from week 8 to 14 fully prevented the reductions in aortic miR-145 and attenuated by 50% the decreases in α-actin, despite uremia-induced hyperphosphatemia. In conclusion, vitamin D was able to prevent the reductions in aortic miR-145 and α-actin content induced by uremia, reducing the alterations in vascular contractility and osteogenic differentiation despite hyperphosphatemia.


Subject(s)
Hyperphosphatemia , MicroRNAs , Uremia , Vascular Calcification , Actins , Animals , Core Binding Factor Alpha 1 Subunit/genetics , Mice , MicroRNAs/genetics , Myocytes, Smooth Muscle , Osteogenesis/genetics , Rats , Vascular Calcification/etiology , Vascular Calcification/prevention & control , Vitamin D/adverse effects
5.
Front Med (Lausanne) ; 8: 790513, 2021.
Article in English | MEDLINE | ID: mdl-34950686

ABSTRACT

Chronic Kidney Disease (CKD), a disorder that affects 11% of the world's population, is characterized by an acceleration in skeletal, immune, renal, and cardiovascular aging that increases the risk of cardiovascular mortality by 10- to 20-fold, compared to that in individuals with normal renal function. For more than two decades, the progressive impairment in renal capacity to maintain normal circulating levels of the hormonal form of vitamin D (1,25-dihydroxyvitamin D or calcitriol) was considered the main contributor to the reduced survival of CKD patients. Accordingly, calcitriol administration was the treatment of choice to attenuate the progression of secondary hyperparathyroidism (SHPT) and its adverse impact on bone health and vascular calcification. The development of calcitriol analogs, designed to mitigate the resistance to calcitriol suppression of PTH associated with CKD progression, demonstrated survival benefits unrelated to the control of SHPT or skeletal health. The exhaustive search for the pathophysiology behind survival benefits associated with active vitamin D analogs has identified novel anti-inflammatory, anti-hypertensive, anti-aging actions of the vitamin D endocrine system. A major paradigm shift regarding the use of calcitriol or active vitamin D analogs to improve survival in CKD patients emerged upon demonstration of a high prevalence of vitamin D (not calcitriol) deficiency at all stages of CKD and, more significantly, that maintaining serum levels of the calcitriol precursor, 25(OH)vitamin D, above 23 ng/ml delayed CKD progression. The cause of vitamin D deficiency in CKD, however, is unclear since vitamin D bioactivation to 25(OH)D occurs mostly at the liver. Importantly, neither calcitriol nor its analogs can correct vitamin D deficiency. The goals of this chapter are to present our current understanding of the pathogenesis of vitamin D deficiency in CKD and of the causal link between defective vitamin D bioactivation to calcitriol and the onset of molecular pathways that promote CKD progression independently of the degree of SHPT. An understanding of these mechanisms will highlight the need for identification of novel sensitive biomarkers to assess the efficacy of interventions with vitamin D and/or calcitriol(analogs) to ameliorate CKD progression in a PTH-independent manner.

6.
Nutrients ; 13(8)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34444716

ABSTRACT

BACKGROUND: In COVID-19 patients, low serum vitamin D (VD) levels have been associated with severe acute respiratory failure and poor prognosis. In regular hemodialysis (HD) patients, there is VD deficiency and markedly reduced calcitriol levels, which may predispose them to worse outcomes of COVID-19 infection. Some hemodialysis patients receive treatment with drugs for secondary hyperparathyroidism, which have well known pleiotropic effects beyond mineral metabolism. The aim of this study was to evaluate the impact of VD status and the administration of active vitamin D medications, used to treat secondary hyperparathyroidism, on survival in a cohort of COVID-19 positive HD patients. METHODS: A cross-sectional retrospective observational study was conducted from 12 March to 21 May 2020 in 288 HD patients with positive PCR for SARS-CoV2. Patients were from 52 different centers in Spain. RESULTS: The percent of HD patients with COVID-19 was 6.1% (288 out of 4743). Mortality rate was 28.4% (81/285). Three patients were lost to follow-up. Serum 25(OH)D (calcidiol) level was 17.1 [10.6-27.5] ng/mL and was not significantly associated to mortality (OR 0.99 (0.97-1.01), p = 0.4). Patients receiving active vitamin D medications (16/94 (17%) vs. 65/191(34%), p = 0.003), including calcimimetics (4/49 (8.2%) vs. 77/236 (32.6%), p = 0.001), paricalcitol or calcimimetics (19/117 (16.2%) vs. 62/168 (36.9%); p < 0.001), and also those on both paricalcitol and calcimimetics, to treat secondary hyperparathyroidism (SHPTH) (1/26 (3.8%) vs. 80/259 (30.9%), p < 0.001) showed a lower mortality rate than patients receiving no treatment with either drug. Multivariate Cox regression analysis confirmed this increased survival. CONCLUSIONS: Our findings suggest that the use of paricalcitol, calcimimetics or the combination of both, seem to be associated with the improvement of survival in HD patients with COVID-19. No correlation was found between serum VD levels and prognosis or outcomes in HD patients with COVID-19. Prospective studies and clinical trials are needed to support these findings.


Subject(s)
COVID-19/mortality , Calcitriol/administration & dosage , Ergocalciferols/administration & dosage , Renal Dialysis/mortality , Aged , Aged, 80 and over , COVID-19/blood , Calcifediol/blood , Calcium/blood , Cross-Sectional Studies , Female , Humans , Hyperparathyroidism, Secondary/blood , Hyperparathyroidism, Secondary/drug therapy , Male , Retrospective Studies , SARS-CoV-2/isolation & purification , Survival Analysis , Vitamin D/blood , Vitamin D Deficiency/blood , Vitamin D Deficiency/drug therapy , Vitamin D Deficiency/mortality , Vitamin D Deficiency/virology
7.
Front Med (Lausanne) ; 8: 618286, 2021.
Article in English | MEDLINE | ID: mdl-34113627

ABSTRACT

Background and Aims: Alterations in novel immune cell subsets, such as angiogenic T cells (Tang), senescent T cells (CD4+CD28null), and monocyte subsets are associated with impaired vascular homeostasis in several inflammatory conditions. However, mediators underlying vascular deterioration in chronic kidney disease (CKD) are poorly characterized. This study assessed their role in the vascular deterioration of CKD using a broad spectrum of surrogate markers ranging from altered functionality to overt calcification. Methods: Tang (CD3+CD31+CXCR4+), CD4+CD28null cells, and monocytes [CD14/CD16 subsets and angiotensin-converting enzyme (ACE) expression] were measured in peripheral blood by flow cytometry in 33 CKD stage 5 patients undergoing peritoneal dialysis (CKD5-PD) and 15 healthy controls (HCs). Analyses were replicated in a hemodialysis cohort. Vascular surrogate markers (including adventitial vasa vasorum, pulse wave velocity, intima-media thickness, and vascular calcification) were assessed by appropriate imaging methods. Results: In CKD5-PD, decreased Tang levels (p < 0.001) were unrelated to clinical features or traditional cardiovascular (CV) risk factors but correlated negatively with troponin T levels (r = -0.550, p = 0.003). Instead, CD4+CD28null frequency was increased (p < 0.001), especially in those with vascular calcifications. Quantitative and qualitative differences were also observed within the monocyte pool, a shift toward CD16+ subsets and ACE expression being found in CKD. Equivalent results were observed in the replication cohort. Each subset associated distinctly with adverse vascular outcomes in univariate and multivariate analyses: while Tang depletion was linked to poor vascular function and subclinical atherosclerosis, increases in CD4+CD28null were associated with overt vascular thickening and calcification. Monocytes were not independently associated with vascular outcomes in CKD patients. Conclusions: Novel T cell and monocyte subsets are altered in CKD. Altered T-cell subpopulations, but not monocytes, exhibited distinct associations with different vascular outcomes in CKD. Tang are emerging biomarkers of subclinical vascular deterioration in CKD.

8.
Nephrol Dial Transplant ; 36(5): 793-803, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33416889

ABSTRACT

BACKGROUND: In chronic kidney disease, the activation of the renin-angiotensin-aldosterone system (RAAS) and renal inflammation stimulates renal fibrosis and the progression to end-stage renal disease. The low levels of vitamin D receptor (VDR) and its activators (VDRAs) contribute to worsen secondary hyperparathyroidism and renal fibrosis. METHODS: The 7/8 nephrectomy model of experimental chronic renal failure (CRF) was used to examine the anti-fibrotic effects of treatment with two VDRAs, paricalcitol and calcitriol, at equivalent doses (3/1 dose ratio) during 4 weeks. RESULTS: CRF increased the activation of the RAAS, renal inflammation and interstitial fibrosis. Paricalcitol treatment reduced renal collagen I and renal interstitial fibrosis by decreasing the activation of the RAAS through renal changes in renin, angiotensin receptor 1 (ATR1) and ATR2 mRNAs levels and renal inflammation by decreasing renal inflammatory leucocytes (CD45), a desintegrin and metaloproteinase mRNA, transforming growth factor beta mRNA and protein, and maintaining E-cadherin mRNA levels. Calcitriol showed similar trends without significant changes in most of these biomarkers. CONCLUSIONS: Paricalcitol effectively attenuated the renal interstitial fibrosis induced by CRF through a combination of inhibitory actions on the RAAS, inflammation and epithelial/mesenchymal transition.


Subject(s)
Calcitriol , Animals , Biomarkers/metabolism , Calcitriol/pharmacology , Ergocalciferols , Fibrosis , Hyperparathyroidism, Secondary/drug therapy , Inflammation/metabolism , Kidney/metabolism , Kidney Failure, Chronic/complications , Receptors, Calcitriol/metabolism , Renal Insufficiency, Chronic/complications , Renin/metabolism , Renin-Angiotensin System/drug effects
9.
Nephrol Dial Transplant ; 36(4): 618-631, 2021 03 29.
Article in English | MEDLINE | ID: mdl-33367746

ABSTRACT

BACKGROUND: In chronic kidney disease, serum phosphorus (P) elevations stimulate parathyroid hormone (PTH) production, causing severe alterations in the bone-vasculature axis. PTH is the main regulator of the receptor activator of nuclear factor κB (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system, which is essential for bone maintenance and also plays an important role in vascular smooth muscle cell (VSMC) calcification. The discovery of a new RANKL receptor, leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4), which is important for osteoblast differentiation but with an unknown role in vascular calcification (VC), led us to examine the contribution of LGR4 in high P/high PTH-driven VC. METHODS: In vivo studies were conducted in subtotally nephrectomized rats fed a normal or high P diet, with and without parathyroidectomy (PTX). PTX rats were supplemented with PTH(1-34) to achieve physiological serum PTH levels. In vitro studies were performed in rat aortic VSMCs cultured in control medium, calcifying medium (CM) or CM plus 10-7 versus 10-9 M PTH. RESULTS: Rats fed a high P diet had a significantly increased aortic calcium (Ca) content. Similarly, Ca deposition was higher in VSMCs exposed to CM. Both conditions were associated with increased RANKL and LGR4 and decreased OPG aorta expression and were exacerbated by high PTH. Silencing of LGR4 or parathyroid hormone receptor 1 (PTH1R) attenuated the high PTH-driven increases in Ca deposition. Furthermore, PTH1R silencing and pharmacological inhibition of protein kinase A (PKA), but not protein kinase C, prevented the increases in RANKL and LGR4 and decreased OPG. Treatment with PKA agonist corroborated that LGR4 regulation is a PTH/PKA-driven process. CONCLUSIONS: High PTH increases LGR4 and RANKL and decreases OPG expression in the aorta, thereby favouring VC. The hormone's direct pro-calcifying actions involve PTH1R binding and PKA activation.


Subject(s)
Myocytes, Smooth Muscle/metabolism , Osteoprotegerin/metabolism , Parathyroid Hormone/pharmacology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, G-Protein-Coupled/metabolism , Vascular Calcification/metabolism , Animals , Calcium-Regulating Hormones and Agents/pharmacology , Gene Expression Regulation/drug effects , Ligands , Male , NF-kappa B/metabolism , Osteoprotegerin/genetics , RANK Ligand/genetics , Rats , Rats, Wistar , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptors, G-Protein-Coupled/genetics
10.
Sci Rep ; 9(1): 17810, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31780737

ABSTRACT

In chronic kidney disease (CKD), hyperphosphatemia-induced inflammation aggravates vascular calcification (VC) by increasing vascular smooth muscle cell (VSMC) osteogenic differentiation, ADAM17-induced renal and vascular injury, and TNFα-induction of neutral-sphingomyelinase2 (nSMase2) to release pro-calcifying exosomes. This study examined anti-inflammatory ß-glucans efficacy at attenuating systemic inflammation in health, and renal and vascular injury favoring VC in hyperphosphatemic CKD. In healthy adults, dietary barley ß-glucans (Bßglucans) reduced leukocyte superoxide production, inflammatory ADAM17, TNFα, nSMase2, and pro-aging/pro-inflammatory STING (Stimulator of interferon genes) gene expression without decreasing circulating inflammatory cytokines, except for γ-interferon. In hyperphosphatemic rat CKD, dietary Bßglucans reduced renal and aortic ADAM17-driven inflammation attenuating CKD-progression (higher GFR and lower serum creatinine, proteinuria, kidney inflammatory infiltration and nSMase2), and TNFα-driven increases in aortic nSMase2 and calcium deposition without improving mineral homeostasis. In VSMC, Bßglucans prevented LPS- or uremic serum-induced rapid increases in ADAM17, TNFα and nSMase2, and reduced the 13-fold higher calcium deposition induced by prolonged calcifying conditions by inhibiting osteogenic differentiation and increases in nSMase2 through Dectin1-independent actions involving Bßglucans internalization. Thus, dietary Bßglucans inhibit leukocyte superoxide production and leukocyte, renal and aortic ADAM17- and nSMase2 gene expression attenuating systemic inflammation in health, and renal injury and aortic calcification despite hyperphosphatemia in CKD.


Subject(s)
ADAM17 Protein/antagonists & inhibitors , Hordeum/chemistry , Renal Insufficiency, Chronic/diet therapy , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Vascular Calcification/diet therapy , beta-Glucans/therapeutic use , Adult , Animals , Disease Models, Animal , Female , Healthy Volunteers , Humans , Inflammation/diet therapy , Male , Mice , Middle Aged , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Young Adult , beta-Glucans/pharmacology
11.
Sci Rep ; 9(1): 13230, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31519925

ABSTRACT

Inflammation is central to chronic kidney disease (CKD) pathogenesis and vascular outcomes, but the exact players remain unidentified. Since low density granulocytes (LDGs) are emerging mediators in inflammatory conditions, we aimed to evaluate whether LDGs may be altered in CKD and related to clinical outcomes as biomarkers. To his end, LDGs subsets were measured in peripheral blood by flow cytometry and confocal microscopy in 33 CKD patients undergoing peritoneal dialysis and 15 healthy controls (HC). Analyses were replicated in an additional cohort. DEF3 (marker of early granulopoiesis) gene expression on PBMCs was quantified by qPCR. Total CD15+ LDGs and both CD14lowCD16+ and CD14-CD16- subsets were expanded in CKD. The relative frequency of the CD14-CD16- subpopulation was higher among the CD15+ pool in CKD. This alteration was stable over-time. The increased CD14-CD16-CD15+ paralleled Kauppila scores and DEF3 expression, whereas no association was found with CD14lowCD16+ CD15+. Both subsets differed in their CD11b, CD10, CD35, CD31, CD62L, IFNAR1 and CD68 expression, FSC/SSC features and nuclear morphology, pointing to different origins and maturation status. In conclusion, LDGs were expanded in CKD showing a skewed distribution towards a CD14-CD16-CD15+ enrichment, in association with vascular calcification. DEF3 expression in PBMC can be a marker of LDG expansion.


Subject(s)
Biomarkers/analysis , Granulocytes/pathology , Inflammation/complications , Renal Insufficiency, Chronic/pathology , Vascular Calcification/complications , Adult , Aged , Female , Humans , Leukocytes, Mononuclear/pathology , Male , Middle Aged , Monocytes/pathology , Neutrophils/pathology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/etiology , Young Adult
12.
Nephrol Dial Transplant ; 34(6): 934-941, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30189026

ABSTRACT

BACKGROUND: In chronic kidney disease (CKD), increases in serum phosphate and parathyroid hormone (PTH) aggravate vascular calcification (VC) and bone loss. This study was designed to discriminate high phosphorus (HP) and PTH contribution to VC and bone loss. METHODS: Nephrectomized rats fed a HP diet underwent either sham operation or parathyroidectomy and PTH 1-34 supplementation to normalize serum PTH. RESULTS: In uraemic rats fed a HP diet, parathyroidectomy with serum PTH 1-34 supplementation resulted in (i) reduced aortic calcium (80%) by attenuating osteogenic differentiation (higher α-actin; reduced Runx2 and BMP2) and increasing the Wnt inhibitor Sclerostin, despite a similar degree of hyperphosphataemia, renal damage and serum Klotho; (ii) prevention of bone loss mostly by attenuating bone resorption and increases in Wnt inhibitors; and (iii) a 70% decrease in serum calcitriol levels despite significantly reduced serum Fgf23, calcium and renal 24-hydroxylase, which questions that Fgf23 is the main regulator of renal calcitriol production. Significantly, when vascular smooth muscle cells (VSMCs) were exposed exclusively to high phosphate and calcium, high PTH enhanced while low PTH attenuated calcium deposition through parathyroid hormone 1 receptor (PTH1R) signalling. CONCLUSIONS: In hyperphosphataemic CKD, a defective suppression of high PTH exacerbates HP-mediated osteogenic VSMC differentiation and reduces vascular levels of anti-calcifying sclerostin.


Subject(s)
Parathyroid Hormone/blood , Phosphates/blood , Renal Insufficiency, Chronic/blood , Vascular Calcification/metabolism , Animals , Bone Diseases, Metabolic/blood , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Proteins/metabolism , Calcitriol/blood , Calcium/blood , Calcium/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Genetic Markers , Hyperphosphatemia/metabolism , Kidney/drug effects , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nephrectomy , Osteogenesis/drug effects , Parathyroid Hormone/therapeutic use , Parathyroidectomy , Phosphorylation , Rats , Rats, Wistar , Vitamin D3 24-Hydroxylase/metabolism
13.
Kidney Int ; 90(1): 77-89, 2016 07.
Article in English | MEDLINE | ID: mdl-27165819

ABSTRACT

Bone loss and increased fractures are common complications in chronic kidney disease. Because Wnt pathway activation is essential for normal bone mineralization, we assessed whether Wnt inhibition contributes to high-phosphorus-induced mineralization defects in uremic rats. By week 20 after 7/8 nephrectomy, rats fed a high-phosphorus diet had the expected high serum creatinine, phosphorus, parathyroid hormone, and fibroblast growth factor 23 (FGF23) levels and low serum calcium. There was a 15% reduction in tibial mineral density and a doubling of bone cortical porosity compared to uremic rats fed a normal-phosphorus diet. The decreases in tibial mineral density were preceded by time-dependent increments in gene expression of bone formation (Osteocalcin and Runx2) and resorption (Cathepsin K) markers, which paralleled elevations in gene expression of the Wnt inhibitors Sfrp1 and Dkk1 in bone. Similar elevations of Wnt inhibitors plus an increased phospho-ß-catenin/ß-catenin ratio occurred upon exposure of the osteoblast cell line UMR106-01 either to uremic serum or to the combination of parathyroid hormone, FGF23, and soluble Klotho, at levels present in uremic serum. Strikingly, while osteoblast exposure to parathyroid hormone suppressed the expression of Wnt inhibitors, FGF23 directly inhibited the osteoblastic Wnt pathway through a soluble Klotho/MAPK-mediated process that required Dkk1 induction. Thus, the induction of Dkk1 by FGF23/soluble Klotho in osteoblasts inactivates Wnt/ß-catenin signaling. This provides a novel autocrine/paracrine mechanism for the adverse impact of high FGF23 levels on bone in chronic kidney disease.


Subject(s)
Decalcification, Pathologic/metabolism , Fibroblast Growth Factors/metabolism , Osteoblasts/metabolism , Renal Insufficiency, Chronic/complications , Wnt Signaling Pathway , Animals , Biomarkers/blood , Biomarkers/metabolism , Calcification, Physiologic , Calcium/blood , Cathepsin K/metabolism , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/metabolism , Decalcification, Pathologic/etiology , Disease Models, Animal , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/pharmacology , Glucuronidase/metabolism , Glucuronidase/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Klotho Proteins , Male , Membrane Proteins/metabolism , Osteoblasts/drug effects , Osteocalcin/metabolism , Parathyroid Hormone/blood , Phosphorus/blood , Phosphorus/metabolism , Phosphorus, Dietary/adverse effects , Porosity , Rats , Rats, Wistar , Renal Insufficiency, Chronic/metabolism , Tibia/metabolism , Tibia/pathology , Uremia/complications , Uremia/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/blood
14.
Clin Kidney J ; 8(4): 456-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26251717

ABSTRACT

This case report presents fluoconazole efficacy to reduce hypercalcaemia and increased urinary calcium excretion in a patient with nephrocalcinosis after a long history of recurrent renal stones caused by a loss-of-function mutation of the CYP24A1 gene. The CYP24A1 gene codes for a key enzyme in the vitamin D endocrine system that protects against vitamin D toxicity by degrading the circulating excess of both 1,25-dihydroxyvitamin D, the hormonal form of vitamin D, and its precursor, 25-hydroxyvitamin D. In order to expedite the identification of this rare disorder and improve therapies to avoid its progression to nephrocalcinosis, this editorial updates the current knowledge on the frequency of CYP24A1-inactivating mutations, the features of their early clinical presentation and progression, and the pathophysiology of vitamin D activation in health and in granulomatous disorders that may help improve current treatment.

15.
Biomed Res Int ; 2015: 604275, 2015.
Article in English | MEDLINE | ID: mdl-25815325

ABSTRACT

Vitamin D deficiency is associated with a range of clinical disorders. To study the mechanisms involved and improve treatments, animal models are tremendously useful. Current vitamin D deficient rat models have important practical limitations, including time requirements when using, exclusively, a vitamin D deficient diet. More importantly, induction of hypovitaminosis D causes significant fluctuations in parathyroid hormone (PTH) and mineral levels, complicating the interpretation of study results. To overcome these shortcomings, we report the successful induction of vitamin D deficiency within three weeks, with stable serum PTH and minerals levels, in Wistar rats. We incorporated two additional manoeuvres compared to a conventional diet. Firstly, the vitamin D depleted diet is calcium (Ca) enriched, to attenuate the development of secondary hyperparathyroidism. Secondly, six intraperitoneal injections of paricalcitol during the first two weeks are given to induce the rapid degradation of circulating vitamin D metabolites. After three weeks, serum 25-hydroxyvitamin D3 (25D) and 1,25-dihydroxyvitamin D3 (1,25D) levels had dropped below detection limits, with unchanged serum PTH, Ca, and phosphate (P) levels. Therefore, this model provides a useful tool to examine the sole effect of hypovitaminosis D, in a wide range of research settings, without confounding changes in PTH, Ca, and P.


Subject(s)
Calcium/administration & dosage , Ergocalciferols/administration & dosage , Vitamin D Deficiency/blood , Vitamin D/blood , Animals , Dietary Supplements , Disease Models, Animal , Humans , Hyperparathyroidism/blood , Minerals/blood , Parathyroid Hormone/blood , Rats , Vitamin D/administration & dosage , Vitamin D Deficiency/chemically induced , Vitamin D Deficiency/pathology
16.
Lab Invest ; 94(6): 608-22, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24732451

ABSTRACT

Vitamin D (VD) deficiency has been suggested as a risk factor for cancer. One recognized mechanism is that the low-serum 25-hydroxyvitamin D (25(OH)D) of VD deficiency reduces intratumoral 25(OH)D conversion to 1α,25-dihydroxyvitamin D (1,25D, the hormonal form of VD), compromising 1,25D-VD receptor (VDR) antitumoral actions. Reduced tumoral VDR and increased CYP24A1, the enzyme that degrades 1,25D and 25(OH)D, further worsen cancer progression. Importantly, in cells expressing CYP27A1 and/or CYP2R1, which convert inert VD into 25(OH)D, low-serum VD may reduce intratumoral 25(OH)D synthesis thereby compromising VDR antitumoral actions because 25(OH)D can activate the VDR directly and enhance 1,25D-VDR action. Therefore, this study examined whether abnormal endometrial expression of CYP27A1 and/or CYP2R1 may impair VDR-antiproliferative properties in endometrial carcinoma (EC). Immunohistochemical analysis of tissue microarrays of normal human endometrium (NE; n=60) and EC (n=157) showed the expected lower VDR expression in EC (P=0.0002). Instead, CYP24A1 expression was lower in EC compared with NE, while CYP27A1 and CYP2R1 expressions were higher (P=0.0002; P=0.03). Furthermore, in NE and EC, CYP2R1 and CYP27A1 expression correlated directly with nuclear VDR levels, an indicator of ligand-induced VDR activation, and inversely with the proliferation marker Ki67. Accordingly, in the endometrioid carcinoma cell lines IK, RL95/2 and HEC1-A, which express VDR, CYP27A1, and CYP2R1, VD efficaciously reduced cell viability and colony number, with a time course that paralleled actual increases in both intracellular 25(OH)D and nuclear VDR levels. Thus, VD may protect from EC progression in part through increased intratumoral 25(OH)D production by CYP27A1 and CYP2R1 for autocrine/paracrine enhancement of 1,25D-VDR-antiproliferative actions.


Subject(s)
Cholecalciferol/pharmacology , Cholestanetriol 26-Monooxygenase/metabolism , Endometrial Neoplasms/chemistry , Endometrium/chemistry , Receptors, Calcitriol/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Line, Tumor , Cell Proliferation/drug effects , Cholestanetriol 26-Monooxygenase/analysis , Cytochrome P450 Family 2 , Female , Humans , Middle Aged , Receptors, Calcitriol/analysis
17.
Curr Vasc Pharmacol ; 12(2): 272-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23713878

ABSTRACT

The integrity of the vitamin D endocrine system is essential for human health. Nutritional vitamin D deficiency in otherwise healthy individuals, associates with a higher risk of mortality for all causes, despite normal serum calcitriol. These deadly causes extend beyond the recognized adverse impact of vitamin D deficiency on calcium and phosphate homeostasis predisposing to secondary hyperparathyroidism, bone loss and vascular calcification. Vitamin D deficiency also associates with an early onset of disorders of aging, including hypertension, proteinuria, insulin resistance, immune abnormalities that enhance the propensity for viral and bacterial infections, autoimmune disorders, cancer, and multiple organ damage due to excessive systemic inflammation causing atherosclerosis, vascular stiffness, renal lesions, and impaired DNA-damage responses. The frequency and severity of all of these disorders markedly increase in chronic kidney disease (CKD) because the kidney is essential to maintain serum levels of calcitriol, the most potent endogenous endocrine activator of the vitamin D receptor (VDR), and also of 25-hydroxyvitamin D, for local rather than systemic VDR activation. The goal of this review is to update the current understanding of the pathophysiology behind the classical and non-classical actions of VDR activation that help prevent the onset and/or attenuate the progression of renal and cardiovascular damage in CKD. This knowledge is essential to identify non-invasive, sensitive and accurate biomarkers of the severity of these disorders, a first step to generate evidence-based recommendations for a safe correction of vitamin D and/or calcitriol deficiency in the course of CKD that effectively improves outcomes.


Subject(s)
Endocrine System , Vitamin D/physiology , Animals , DNA Damage , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/physiology , Humans , Proteinuria/prevention & control , Receptors, Calcitriol/physiology , Renal Insufficiency, Chronic/etiology , Renin-Angiotensin System/physiology
18.
Kidney Int ; 82(10): 1046-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23128114

ABSTRACT

In normal people, induction by FGF23 of renal Cyp24a1, the enzyme that degrades 25-hydroxyvitamin D and 1,25-dihydroxyvitamin D, helps to protect from the disorders caused by excessive 1,25-dihydroxyvitamin D. Dai and co-workers report, however, that in human and mouse kidney disease, high FGF23 concurs with low rather than high serum 24,25-dihydroxyvitamin D, a biomarker of Cyp24a1 activity. Their characterization of the underlying mechanisms provides new understanding of how kidney disease impairs the health benefits of vitamin D-FGF23/klotho interactions.


Subject(s)
Dihydroxycholecalciferols/blood , Fibroblast Growth Factors/blood , Nephritis, Hereditary/blood , Renal Insufficiency, Chronic/blood , Animals , Female , Fibroblast Growth Factor-23 , Humans , Male
19.
Kidney Int ; 81(10): 937-939, 2012 May.
Article in English | MEDLINE | ID: mdl-22543902

ABSTRACT

Therapy to enhance the renoprotective actions of vitamin D receptor (VDR) activation should safely overcome the distinct VDR content along the nephron to effectively control renal calcium reabsorption, control renal klotho levels for the phosphaturic actions of FGF23, and inhibit proteinuria and the activation of the renin-angiotensin system.


Subject(s)
Kidney/chemistry , Receptors, Calcitriol/analysis , Animals , Female , Fibroblast Growth Factor-23 , Humans , Male
20.
Kidney Int ; 79(7): 715-29, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21270766

ABSTRACT

In kidney disease, the progressive loss of renal capacity to produce calcitriol, the vitamin D hormone, is a key contributor to elevations in parathyroid hormone (PTH) and mineral and skeletal disorders predisposing to renal and cardiovascular damage, ectopic calcifications, and high mortality rates. Thus, the safe correction of calcitriol deficiency to suppress PTH has been the treatment of choice for decades. However, recent epidemiological and experimental data suggest that calcitriol replacement may improve outcomes through renal and cardioprotective actions unrelated to PTH suppression. Furthermore, a striking incidence of vitamin D deficiency occurs in kidney disease and associates more strongly than calcitriol deficiency with a higher risk for kidney disease progression and death. Despite the translational relevance of these findings, no prospective trials are currently available in support of the efficacy of vitamin D supplementation and/or calcitriol replacement to safely halt/moderate renal disease progression. This review updates the pathophysiology behind the vicious cycle by which kidney injury impairs the maintenance of normal vitamin D and calcitriol levels, which in turn impedes vitamin D/calcitriol renoprotective actions, a requirement for the design of prospective trials to improve current recommendations for vitamin D interventions at all stages of kidney disease.


Subject(s)
Kidney Diseases/metabolism , Kidney/metabolism , Signal Transduction , Vitamin D/metabolism , Animals , Calcitriol/therapeutic use , Dietary Supplements , Disease Progression , Humans , Kidney/drug effects , Kidney/physiopathology , Kidney Diseases/complications , Kidney Diseases/drug therapy , Kidney Diseases/physiopathology , Practice Guidelines as Topic , Receptors, Calcitriol/metabolism , Signal Transduction/drug effects , Vitamin D/analogs & derivatives , Vitamin D/blood , Vitamin D/therapeutic use , Vitamin D Deficiency/complications , Vitamin D Deficiency/drug therapy , Vitamin D Deficiency/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...