Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Kidney Int ; 104(4): 698-706, 2023 10.
Article in English | MEDLINE | ID: mdl-37541585

ABSTRACT

Vitamin D supplements have long been advocated for people with chronic kidney disease based on data from observational studies among the general population and people with chronic kidney disease. These data consistently suggested that higher circulating concentrations of 25-hydroxyvitamin D are associated with improved fracture, cardiovascular, cancer, and mortality outcomes. In the past few years, large clinical trials have been conducted to assess the effects of vitamin D supplements on a range of clinically relevant outcomes. Most of these studies were performed in the general population, but they also enrolled people with chronic kidney disease. Virtually all of these trials were negative and contradicted the observational data. In this review, the key observational data and clinical trials are summarized, and potential explanations for the discrepancies between these studies are discussed.


Subject(s)
Renal Insufficiency, Chronic , Vitamin D , Humans , Dietary Supplements , Fractures, Bone , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Vitamin D/therapeutic use , Observational Studies as Topic
2.
Nutrients ; 15(3)2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36771254

ABSTRACT

Awareness of the clinical relevance of magnesium in medicine has increased over the last years, especially for people with chronic kidney disease (CKD), due to magnesium's role in vascular calcification and mineral metabolism. The inverse association between serum magnesium and clinically relevant, adverse outcomes is well-established in people with CKD. Subsequent intervention studies have focused on the effect of magnesium administration, mainly in relation to cardiovascular diseases, mineral bone metabolism, and other metabolic parameters. The most commonly used routes of magnesium administration are orally and by increasing dialysate magnesium. Several oral magnesium formulations are available and the daily dosage of elemental magnesium varies highly between studies, causing considerable heterogeneity. Although data are still limited, several clinical studies demonstrated that magnesium administration could improve parameters of vascular function and calcification and mineral metabolism in people with CKD. Current clinical research has shown that magnesium administration in people with CKD is safe, without concerns for severe hypermagnesemia or negative interference with bone metabolism. It should be noted that there are several ongoing magnesium intervention studies that will contribute to the increasing knowledge on the potential of magnesium administration in people with CKD.


Subject(s)
Cardiovascular Diseases , Renal Insufficiency, Chronic , Vascular Calcification , Humans , Magnesium , Renal Insufficiency, Chronic/complications , Cardiovascular Diseases/etiology , Minerals , Vascular Calcification/complications
3.
Trials ; 23(1): 769, 2022 Sep 12.
Article in English | MEDLINE | ID: mdl-36096824

ABSTRACT

BACKGROUND: Arterial stiffness and calcification propensity are associated with high cardiovascular risk and increased mortality in chronic kidney disease (CKD). Both magnesium and phosphate are recognized as modulators of vascular calcification and chronic inflammation, both features of CKD that contribute to arterial stiffness. In this paper, we outline the rationale and design of a randomized controlled trial (RCT) investigating whether 24 weeks of oral magnesium supplementation with or without additional phosphate-binding therapy can improve arterial stiffness and calcification propensity in patients with stage 3-4 CKD. METHODS: In this multi-center, placebo-controlled RCT, a total of 180 participants with an estimated glomerular filtration rate of 15 to 50 ml/min/1.73 m2 without phosphate binder therapy will be recruited. During the 24 weeks intervention, participants will be randomized to one of four intervention groups to receive either magnesium citrate (350 mg elemental magnesium/day) or placebo, with or without the addition of the phosphate binder sucroferric oxyhydroxide (1000 mg/day). Primary outcome of the study is the change of arterial stiffness measured by the carotid-femoral pulse wave velocity over 24 weeks. Secondary outcomes include markers of calcification and inflammation, among others calcification propensity (T50) and high-sensitivity C-reactive protein. As explorative endpoints, repeated 18F-FDG and 18F-NaF PET-scans will be performed in a subset of participants (n = 40). Measurements of primary and secondary endpoints are performed at baseline, 12 and 24 weeks. DISCUSSION: The combined intervention of magnesium citrate supplementation and phosphate-lowering therapy with sucroferric oxyhydroxide, in stage 3-4 CKD patients without overt hyperphosphatemia, aims to modulate the complex and deregulated mineral metabolism leading to vascular calcification and arterial stiffness and to establish to what extent this is mediated by T50 changes. The results of this combined intervention may contribute to future early interventions for CKD patients to reduce the risk of CVD and mortality. TRIAL REGISTRATION: Netherlands Trial Register, NL8252 (registered December 2019), EU clinical Trial Register 2019-001306-23 (registered November 2019).


Subject(s)
Renal Insufficiency, Chronic , Vascular Calcification , Vascular Diseases , Vascular Stiffness , Citric Acid , Dietary Supplements/adverse effects , Humans , Inflammation , Magnesium/adverse effects , Organometallic Compounds , Phosphates , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/drug therapy , Vascular Calcification/complications , Vascular Calcification/diagnostic imaging , Vascular Calcification/drug therapy
4.
Nephrol Dial Transplant ; 37(6): 1049-1058, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35134986

ABSTRACT

BACKGROUND: Vascular calcification is a key process involved in cardiovascular morbidity and mortality in patients with chronic kidney disease (CKD). Magnesium supplementation may counteract vascular calcification. In this study we aimed to determine whether increased dietary magnesium intake inhibits vascular calcification in CKD in vivo and explore the mechanisms underlying these effects. METHODS: Sprague Dawley rats were partially nephrectomized and fed a diet with high phosphate and either high or normal magnesium content for 16 weeks. The primary outcome was the tissue calcium content of the aorta in the high versus normal dietary magnesium group. In addition, we analysed plasma mineral concentrations, aortic vascular calcification identified with von Kossa staining, calcium apposition time and aortic expression of genes related to vascular calcification. RESULTS: The number of animals in the highest tissue calcium content tertile was significantly lower in the abdominal aorta [1 (10%) versus 6 (55%); P = .03] in the high versus normal dietary magnesium group, but did not differ in the aortic arch and thoracic aorta. Von Kossa staining and calcium apposition time corresponded to these results. The median tissue calcium content was not significantly different between the groups. Serum phosphate concentrations and expression of osteogenic markers in the aorta did not differ between the groups. CONCLUSIONS: This study demonstrates that increased dietary magnesium inhibits abdominal vascular calcification in an experimental animal model of CKD in vivo. These are promising results for CKD patients and further study is needed to identify the mechanisms involved and to determine the clinical relevance in patients.


Subject(s)
Arteriosclerosis , Renal Insufficiency, Chronic , Vascular Calcification , Animals , Aorta, Abdominal , Calcium , Dietary Supplements , Disease Models, Animal , Humans , Magnesium/pharmacology , Magnesium/therapeutic use , Models, Animal , Phosphates , Rats , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/drug therapy , Vascular Calcification/etiology , Vascular Calcification/prevention & control
5.
J Nephrol ; 35(3): 875-888, 2022 04.
Article in English | MEDLINE | ID: mdl-35138627

ABSTRACT

Hyperphosphatemia is a common complication in dialysis-dependent patients with chronic kidney disease. Most dialysis-dependent patients need oral phosphate binder therapy to control serum phosphorus concentrations. Most phosphate binders have a high daily pill burden, which may reduce treatment adherence and impair phosphorus control. Sucroferric oxyhydroxide is a potent iron-based phosphate binder approved for use in dialysis-dependent patients in 2013. A randomized controlled trial of sucroferric oxyhydroxide demonstrated its efficacy for reduction of serum phosphorus with a lower pill burden than sevelamer carbonate. Clinical trials carefully select patients, monitor adherence, and routinely titrate medications to a protocol-defined goal. Consequently, trials may not reflect real-world use of medications. Since its approval, we and others have performed retrospective and prospective analyses of sucroferric oxyhydroxide in real-world clinical practice in > 6400 hemodialysis and approximately 500 peritoneal dialysis patients in the USA and Europe. Consistent with the clinical trial data, real-world observational studies have demonstrated that sucroferric oxyhydroxide can effectively reduce serum phosphorus with a lower daily pill burden than most other phosphate binders. These studies have also shown sucroferric oxyhydroxide provides effective serum phosphorus control in different treatment settings, including as monotherapy in phosphate binder-naïve patients, in patients switching from other phosphate binders, or when used in combination with other phosphate binders. These observational studies indicate a favorable safety and tolerability profile, and minimal, if any, systemic iron absorption. This article reviews the key results from these observational studies of sucroferric oxyhydroxide and evaluates its role in the management of hyperphosphatemia in clinical practice.


Subject(s)
Hyperphosphatemia , Drug Combinations , Ferric Compounds/therapeutic use , Humans , Hyperphosphatemia/drug therapy , Hyperphosphatemia/etiology , Iron/therapeutic use , Phosphates , Phosphorus , Prospective Studies , Randomized Controlled Trials as Topic , Renal Dialysis/adverse effects , Retrospective Studies , Sucrose/therapeutic use
6.
Nephrol Dial Transplant ; 37(4): 652-662, 2022 03 25.
Article in English | MEDLINE | ID: mdl-34718756

ABSTRACT

BACKGROUND: Hyperphosphataemia is strongly associated with cardiovascular disease and mortality. Recently, phosphate binders (PBs), which are used to bind intestinal phosphate, have been shown to bind vitamin K, thereby potentially aggravating vitamin K deficiency. This vitamin K binding by PBs may offset the beneficial effects of phosphate reduction in reducing vascular calcification (VC). Here we assessed whether combining PBs with vitamin K2 supplementation inhibits VC. METHODS: We performed 3/4 nephrectomy in rats, after which warfarin was given for 3 weeks to induce vitamin K deficiency. Next, animals were fed a high phosphate diet in the presence of low or high vitamin K2 and were randomized to either control or one of four different PBs for 8 weeks. The primary outcome was the amount of thoracic and abdominal aorta VC measured by high-resolution micro-computed tomography (µCT). Vitamin K status was measured by plasma MK7 levels and immunohistochemically analysed in vasculature using uncarboxylated matrix Gla protein (ucMGP) specific antibodies. RESULTS: The combination of a high vitamin K2 diet and PB treatment significantly reduced VC as measured by µCT for both the thoracic (P = 0.026) and abdominal aorta (P = 0.023), compared with MK7 or PB treatment alone. UcMGP stain was significantly more present in the low vitamin K2-treated groups in both the thoracic (P < 0.01) and abdominal aorta (P < 0.01) as compared with high vitamin K2-treated groups. Moreover, a high vitamin K diet and PBs led to reduced vascular oxidative stress. CONCLUSION: In an animal model of kidney failure with vitamin K deficiency, neither PB therapy nor vitamin K2 supplementation alone prevented VC. However, the combination of high vitamin K2 with PB treatment significantly attenuated VC.


Subject(s)
Renal Insufficiency , Vascular Calcification , Vitamin K Deficiency , Animals , Female , Male , Rats , Calcium-Binding Proteins , Extracellular Matrix Proteins , Models, Animal , Phosphates , Renal Dialysis , Renal Insufficiency/complications , Vascular Calcification/etiology , Vascular Calcification/prevention & control , Vitamin K , Vitamin K 1/therapeutic use , Vitamin K 2/pharmacology , Vitamin K 2/therapeutic use , Vitamin K Deficiency/complications , Vitamin K Deficiency/drug therapy , X-Ray Microtomography
7.
Eur J Nutr ; 60(3): 1645-1654, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32808059

ABSTRACT

OBJECTIVE: To explore the association of both plasma vitamin D and K concentrations with all-cause mortality, cardiovascular mortality, and cardiovascular events in the general population. METHODS: We studied 4742 participants of the Prevention of REnal and Vascular ENd-Stage Disease (PREVEND) Study. At baseline, vitamin D and K status was determined by measurement of 25-hydroxyvitamin D [25(OH)D] and dephosphorylated uncarboxylated matrix Gla protein (dp-ucMGP), respectively. Patients were categorized into: 25(OH)D < 50 or ≥ 50 nmol/L and dp-ucMGP < 361 or ≥ 361 pmol/L with 25(OH)D > 75 nmol/L and dp-ucMGP < 361 pmol/L as reference. Cause of death was coded according to International Classification of Diseases 9&10 codes from the 2001-2003 examination until date of death/event or censoring date (January 1st, 2017). RESULTS: Mean age was 52.6 ± 11.9 years and 2513 (53%) were female. During a median of 14.2 year follow-up, 620 participants died of which 142 were due to cardiovascular causes. Combined low vitamin D and K status was present in 970 participants (20%) and was associated with a greater risk of all-cause mortality compared to high vitamin D and high vitamin K status group (n = 1424) after adjusting for potential confounders: hazard ratio 1.46 (95% confidence intervals 1.12-1.90). We observed similar trends, albeit non-significant for cardiovascular mortality, and cardiovascular events: 1.42 (0.79-2.55), 1.28 (0.93-1.77), respectively. CONCLUSIONS: Combined low vitamin D and K status are associated with increased all-cause mortality risk and possibly with cardiovascular mortality and cardiovascular events compared with adequate vitamin D and K status. Future studies should investigate the effect of combined vitamin D and K supplementation on clinical outcomes.


Subject(s)
Cardiovascular Diseases , Vitamin K Deficiency , Adult , Calcium-Binding Proteins , Female , Humans , Male , Middle Aged , Prospective Studies , Risk Factors , Vitamin D , Vitamin K
8.
Clin Kidney J ; 13(1): 75-84, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32082556

ABSTRACT

BACKGROUND: Etelcalcetide is an intravenous calcimimetic approved for treatment of secondary hyperparathyroidism (sHPT) in patients receiving hemodialysis. Besides lowering parathyroid hormone (PTH), etelcalcetide also significantly reduces fibroblast growth factor 23 (FGF23), but the mechanisms are unknown. METHODS: To investigate potential mediators of etelcalcetide-induced FGF23 reduction, we performed secondary analyses of the 26-week randomized trials that compared the effects on PTH of etelcalcetide (n = 509) versus placebo (n = 514) and etelcalcetide (n = 340) versus cinacalcet (n = 343) in adults with sHPT receiving hemodialysis. We analyzed changes in FGF23 in relation to changes in PTH, calcium, phosphate and bone turnover markers. We also investigated how concomitant treatments aimed at mitigating hypocalcemia altered the FGF23-lowering effects of etelcalcetide. RESULTS: Etelcalcetide reduced FGF23 [median % change (quartile 1-quartile 3)] from baseline to the end of the trial significantly more than placebo [-56% (-85 to -7) versus +2% (-40 to +65); P < 0.001] and cinacalcet [-68% (-87 to -26) versus -41% (-76 to +25); P < 0.001]. Reductions in FGF23 correlated strongly with reductions in calcium and phosphate, but not with PTH; correlations with bone turnover markers were inconsistent and of borderline significance. Increases in concomitant vitamin D administration partially attenuated the FGF23-lowering effect of etelcalcetide, but increased dialysate calcium concentration versus no increase and increased dose of calcium supplementation versus no increase did not attenuate the FGF23-lowering effects of etelcalcetide. CONCLUSION: These data suggest that etelcalcetide potently lowers FGF23 in patients with sHPT receiving hemodialysis and that the effect remains detectable among patients who receive concomitant treatments aimed at mitigating treatment-associated decreases in serum calcium.

9.
Kidney Int ; 97(3): 487-501, 2020 03.
Article in English | MEDLINE | ID: mdl-31866113

ABSTRACT

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


Subject(s)
Glucuronidase/deficiency , Magnesium/pharmacology , Renal Insufficiency, Chronic , Vascular Calcification , Animals , Glucuronidase/genetics , Klotho Proteins , Mice , Mice, Knockout , Parathyroid Hormone , Phosphates , Vascular Calcification/genetics , Vascular Calcification/prevention & control
10.
Nephrol Dial Transplant ; 35(1): 47-64, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31641778

ABSTRACT

Secondary hyperparathyroidism (SHPT) is an important complication of advanced chronic kidney disease (CKD) in children, which is often difficult to treat with conventional therapy. The calcimimetic cinacalcet is an allosteric modulator of the calcium-sensing receptor. It has proven to be effective and safe in adults to suppress parathyroid hormone (PTH), but data on its use in children are limited. To date, studies in children only consist of two randomized controlled trials, nine uncontrolled interventional or observational studies, and case reports that report the efficacy of cinacalcet as a PTH-lowering compound. In 2017, the European Medical Agency approved the use of cinacalcet for the treatment of SHPT in children on dialysis in whom SHPT is not adequately controlled with standard therapy. Since evidence-based guidelines are so far lacking, we present a position statement on the use of cinacalcet in paediatric dialysis patients based on the available evidence and opinion of experts from the European Society for Paediatric Nephrology, Chronic Kidney Disease-Mineral and Bone Disorder and Dialysis Working Groups, and the ERA-EDTA. Given the limited available evidence the strength of these statements are weak to moderate, and must be carefully considered by the treating physician and adapted to individual patient needs as appropriate. Audit and research recommendations to study key outcome measures in paediatric dialysis patients receiving cinacalcet are suggested.


Subject(s)
Calcimimetic Agents/therapeutic use , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Cinacalcet/therapeutic use , Hyperparathyroidism, Secondary/drug therapy , Practice Guidelines as Topic/standards , Renal Dialysis/adverse effects , Child , Chronic Kidney Disease-Mineral and Bone Disorder/therapy , Evidence-Based Medicine , Humans , Hyperparathyroidism, Secondary/etiology
11.
Article in English | MEDLINE | ID: mdl-30805347

ABSTRACT

Calcium supplements are broadly prescribed to treat osteoporosis either as monotherapy or together with vitamin D to enhance calcium absorption. It is still unclear whether calcium supplementation significantly contributes to the reduction of bone fragility and fracture risk. Data suggest that supplementing post-menopausal women with high doses of calcium has a detrimental impact on cardiovascular morbidity and mortality. Chronic kidney disease (CKD) patients are prone to vascular calcification in part due to impaired phosphate excretion. Calcium-based phosphate binders further increase risk of vascular calcification progression. In both bone and vascular tissue, vitamin K-dependent processes play an important role in calcium homeostasis and it is tempting to speculate that vitamin K supplementation might protect from the potentially untoward effects of calcium supplementation. This review provides an update on current literature on calcium supplementation among post-menopausal women and CKD patients and discusses underlying molecular mechanisms of vascular calcification. We propose therapeutic strategies with vitamin K2 treatment to prevent or hold progression of vascular calcification as a consequence of excessive calcium intake.

12.
Nutrients ; 11(2)2019 Jan 22.
Article in English | MEDLINE | ID: mdl-30678199

ABSTRACT

Vitamin D supplementation has been widely promoted to restore 25-hydroxyvitamin D concentrations; however, experimental evidence suggests a nutrient interaction with vitamin K. We assessed the effects of 1200 IU vitamin D3 per day versus placebo for six months on vitamin K status in a randomized, double-blind, placebo-controlled trial with participants aged 60⁻80 years with depressive symptoms and ≥1 functional limitation for a secondary analysis. Stored baseline and six-month follow-up blood samples were available for 131 participants (n = 65 placebo vs. n = 66 vitamin D supplementation). We measured dephosphorylated uncarboxylated matrix gla protein (MGP) (dp-ucMGP) concentrations-a marker of vitamin K deficiency. Mean age was 68 years, and 89 participants (68%) were women. Vitamin K antagonists were used by 16 participants and multivitamin supplements by 50 participants. No differences in change between intervention and placebo were found (-38.5 ± 389 vs. 4.5 ± 127 (pmol/L), p = 0.562). When excluding vitamin K antagonist users and multivitamin users, dp-ucMGP at follow-up was significantly higher in the vitamin D group (n = 40) compared to placebo (n = 30), with a difference of 92.8 (5.7, 180) pmol/L, adjusting for baseline dp-ucMGP and sex. In conclusion, vitamin D supplementation for six months did not affect vitamin K status; however, among participants without vitamin K antagonist or multivitamin use, vitamin D supplementation influenced dp-ucMGP concentrations.


Subject(s)
Calcium-Binding Proteins/blood , Cholecalciferol/pharmacology , Extracellular Matrix Proteins/blood , Aged , Calcium-Binding Proteins/metabolism , Cholecalciferol/administration & dosage , Double-Blind Method , Extracellular Matrix Proteins/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Male , Middle Aged , Matrix Gla Protein
13.
Nephrol Dial Transplant ; 34(2): 252-264, 2019 02 01.
Article in English | MEDLINE | ID: mdl-29718431

ABSTRACT

Background: Uraemia induces endothelial cell (EC) injury and impaired repair capacity, for which the underlying mechanism remains unclear. Active vitamin D (VD) may promote endothelial repair, however, the mechanism that mediates the effects of VD in chronic kidney disease are poorly understood. Thus, we investigated uraemia-induced endothelial damage and the protection against such damage by active VD. Methods: We applied electric cell-substrate impedance sensing (ECIS) to study real-time responses of human ECs exposed to pooled uraemic and non-uraemic plasma with or without the addition of active VD. The effects of indoxyl sulphate and p-cresol were tested in non-uraemic plasma. Structural changes for vascular endothelial (VE)-cadherin and F-actin were assessed by immunostaining and quantified. Results: The exposure of ECs to uraemic media significantly decreased endothelial barrier function after 24 h. Cell migration after electrical wounding and recovery of the barrier after thrombin-induced loss of integrity were significantly impaired in uraemic-medium stimulated cells and cells exposed to indoxyl sulphate and p-cresol. This effect on ECIS was dependent on loss of cell-cell interaction. Mechanistically, we found that EC, exposed to uraemic media, displayed disrupted VE-cadherin interactions and F-actin reorganization. VD supplementation rescued both endothelial barrier function and cell-cell interactions in ECs exposed to uraemic media. These events were associated with an increment of VE-cadherin at intercellular junctions. Conclusions: Our data demonstrate a potentially clinically relevant mechanism for uraemia-induced endothelial damage. Furthermore, active VD rescued the uraemic medium-induced loss of cell-cell adhesion, revealing a novel role of active VD in preservation of endothelial integrity during uraemia.


Subject(s)
Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Uremia/metabolism , Vitamin D/pharmacology , Actins/metabolism , Adult , Aged , Antigens, CD/metabolism , Cadherins/metabolism , Cell Adhesion , Cell Movement , Cells, Cultured , Cresols/pharmacology , Endothelium, Vascular/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Indican/pharmacology , Intercellular Junctions/drug effects , Male , Middle Aged , Thrombin/metabolism , Uremia/drug therapy , Young Adult
14.
J Am Heart Assoc ; 7(17): e008776, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30371149

ABSTRACT

Background Dysfunctional endothelium may contribute to the development of cardiovascular complications in chronic kidney disease ( CKD ). Supplementation with active vitamin D has been proposed to have vasoprotective potential in CKD , not only by direct effects on the endothelium but also by an increment of α-Klotho. Here, we explored the capacity of the active vitamin D analogue paricalcitol to protect against uremia-induced endothelial damage and the extent to which this was dependent on increased α-Klotho concentrations. Methods and Results In a combined rat model of CKD with vitamin D deficiency, renal failure induced vascular permeability and endothelial-gap formation in thoracic aorta irrespective of baseline vitamin D, and this was attenuated by paricalcitol. Downregulation of renal and serum α-Klotho was found in the CKD model, which was not restored by paricalcitol. By measuring the real-time changes of the human endothelial barrier function, we found that paricalcitol effectively improved the recovery of endothelial integrity following the addition of the pro-permeability factor thrombin and the induction of a wound. Furthermore, immunofluorescence staining revealed that paricalcitol promoted vascular endothelial-cadherin-based cell-cell junctions and diminished F-actin stress fiber organization, preventing the formation of endothelial intracellular gaps. Conclusions Our results demonstrate that paricalcitol attenuates the CKD -induced endothelial damage in the thoracic aorta and directly mediates endothelial stability in vitro by enforcing cell-cell interactions.


Subject(s)
Aorta, Thoracic/drug effects , Capillary Permeability/drug effects , Endothelium, Vascular/drug effects , Ergocalciferols/pharmacology , Renal Insufficiency, Chronic/metabolism , Uremia/metabolism , Vitamin D Deficiency/metabolism , Actins/drug effects , Actins/metabolism , Animals , Aorta, Thoracic/metabolism , Cadherins/drug effects , Cadherins/metabolism , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Glucuronidase/drug effects , Glucuronidase/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Klotho Proteins , Rats , Stress Fibers/drug effects
15.
Sci Rep ; 8(1): 5148, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29581540

ABSTRACT

Disturbances in calcium metabolism are common in individuals with chronic kidney disease (CKD), but whether they are associated with subsequent kidney function decline is less clear. In a CKD 3-5 cohort of 15,755 adult citizens of Stockholm with creatinine tests taken during 2006-2011 and concurrent calcium testing at cohort entry, we investigated the association between baseline serum calcium and the subsequent change in estimated glomerular filtration rate (eGFR, by CKD-EPI) decline using linear mixed models. Mean (SD) baseline corrected serum calcium was 9.6 (0.5) mg/dL. Mean (95%-confidence interval [CI]) eGFR decline was -0.82 (-0.90; -0.74) mL/min/1.73 m2/year. In advanced CKD stages, higher baseline serum calcium was associated with less rapid kidney function decline. The adjusted change (95%-CI) in eGFR decline associated with each mg/dL increase in baseline serum calcium was -0.10 (-0.28; 0.26), 0.39 (0.07; 0.71), 0.34 (-0.02; 0.70) and 0.68 (0.36; 1.00) mL/min/1.73 m2/year for individuals in CKD stage 3a, 3b, 4, and 5, respectively. In a subgroup of patients using vitamin D supplements, the association between baseline serum calcium and CKD progression was eliminated, especially in CKD stage 3b and 4. To conclude, in individuals with CKD stage 3b to 5, lower baseline corrected serum calcium, rather than higher baseline serum calcium, associated with a more rapid CKD progression. Lower serum corrected calcium seems to be indicative for vitamin D deficiency.


Subject(s)
Calcium/blood , Disease Progression , Glomerular Filtration Rate , Kidney/physiopathology , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/epidemiology , Aged , Aged, 80 and over , Cohort Studies , Female , Follow-Up Studies , Humans , Linear Models , Male , Middle Aged , Multivariate Analysis , Renal Insufficiency, Chronic/prevention & control , Risk Factors , Sweden/epidemiology , Vitamin D/therapeutic use , Vitamin D Deficiency
16.
Ann Intern Med ; 168(6): 422-430, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29459980

ABSTRACT

Description: The Kidney Disease: Improving Global Outcomes (KDIGO) 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD) is a selective update of the prior CKD-MBD guideline published in 2009. The guideline update and the original publication are intended to assist practitioners caring for adults with CKD and those receiving long-term dialysis. Methods: Development of the guideline update followed an explicit process of evidence review and appraisal. The approach adopted by the Work Group and the evidence review team was based on systematic reviews of relevant trials, appraisal of the quality of the evidence, and rating of the strength of recommendations according to the GRADE (Grading of Recommendations Assessment, Development and Evaluation) approach. Searches of the English-language literature were conducted through September 2015 and were supplemented with targeted searches through February 2017. Final modification of the guidelines was informed by a public review process involving numerous stakeholders, including patients, subject matter experts, and industry and national organizations. Recommendations: The update process resulted in the revision of 15 recommendations. This synopsis focuses primarily on recommendations for diagnosis of and testing for CKD-MBD and treatment of CKD-MBD that emphasizes decreasing phosphate levels, maintaining calcium levels, and addressing elevated parathyroid hormone levels in adults with CKD stage G3a to G5 and those receiving dialysis. Key elements include basing treatment on trends in laboratory values rather than a single abnormal result and being cautious to avoid hypercalcemia when treating secondary hyperparathyroidism.


Subject(s)
Chronic Kidney Disease-Mineral and Bone Disorder/diagnosis , Chronic Kidney Disease-Mineral and Bone Disorder/therapy , Chronic Kidney Disease-Mineral and Bone Disorder/prevention & control , Humans , Hypercalcemia/prevention & control , Hyperphosphatemia/blood , Hyperphosphatemia/prevention & control , Parathyroid Hormone/blood , Renal Dialysis
17.
Kidney Int ; 92(1): 26-36, 2017 07.
Article in English | MEDLINE | ID: mdl-28646995

ABSTRACT

The KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of CKD-MBD represents a selective update of the prior CKD-MBD Guideline published in 2009. This update, along with the 2009 publication, is intended to assist the practitioner caring for adults and children with chronic kidney disease (CKD), those on chronic dialysis therapy, or individuals with a kidney transplant. This review highlights key aspects of the 2017 CKD-MBD Guideline Update, with an emphasis on the rationale for the changes made to the original guideline document. Topic areas encompassing updated recommendations include diagnosis of bone abnormalities in CKD-mineral and bone disorder (MBD), treatment of CKD-MBD by targeting phosphate lowering and calcium maintenance, treatment of abnormalities in parathyroid hormone in CKD-MBD, treatment of bone abnormalities by antiresorptives and other osteoporosis therapies, and evaluation and treatment of kidney transplant bone disease.


Subject(s)
Bone Remodeling/drug effects , Chelating Agents/therapeutic use , Chronic Kidney Disease-Mineral and Bone Disorder/drug therapy , Dietary Supplements , Evidence-Based Medicine/standards , Nephrology/standards , Vitamin D/therapeutic use , Biomarkers/blood , Calcium/blood , Chelating Agents/adverse effects , Chronic Kidney Disease-Mineral and Bone Disorder/blood , Chronic Kidney Disease-Mineral and Bone Disorder/diagnosis , Consensus , Dietary Supplements/adverse effects , Humans , Parathyroid Hormone/blood , Phosphates/blood , Risk Factors , Treatment Outcome , Vitamin D/adverse effects
18.
Magnes Res ; 29(4): 126-140, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-28007671

ABSTRACT

Magnesium (Mg) is one of the most important cations in the body, playing an essential role in biological systems as co-factor for more than 300 essential enzymatic reactions. In the general population, low levels of Mg are associated with a high risk of cardio-vascular disease (CVD). Despite the accumulating literature data, the effect of Mg administration on mortality in chronic kidney disease (CKD) patients has never been investigated as a primary end-point. We conducted a systematic search of studies assessing the benefits and harms of Mg in CKD (stages 1 to 5 and 5D), and considered all randomized controlled trials (RCTs) and quasi-RCTs evaluating Mg-based interventions in CKD. As a phosphate binder, Mg salts offer a plausible opportunity for doubly favorable effects via reduction of intestinal phosphate absorption and addition of potentially beneficial effects via increasing circulating Mg levels. Mg supplementation might have a favorable effect on vascular calcification, although evidence for this is very slight. Although longitudinal data describe an association between low serum Mg levels and increased total and cardiovascular mortality, in patients with CKD, the existing RCTs reporting the effect of Mg supplementation on mortality failed to demonstrate any favorable effect. As with many other variables that influence hard end-points in nephrology, the role of Mg in CKD patients needs to be investigated in more depth. Additional research that is well-designed and directly targeting the role of Mg is needed as a consequence of limited existing evidence.


Subject(s)
Kidney/drug effects , Magnesium/pharmacology , Magnesium/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Animals , Humans , Kidney/metabolism , Magnesium/administration & dosage , Magnesium/adverse effects , Renal Insufficiency, Chronic/metabolism
19.
Kidney Int ; 89(3): 625-35, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26880457

ABSTRACT

Peritoneal dialysis (PD) can result in chronic inflammation and progressive peritoneal membrane damage. Alanyl-Glutamine (Ala-Gln), a dipeptide with immunomodulatory effects, improved resistance of mesothelial cells to PD fluids. Recently, interleukin-17 (IL-17) was found to be associated with PD-induced peritoneal damage. Here we studied the capacity of intraperitoneal Ala-Gln administration to protect against peritoneal damage by modulating IL-17 expression in uremic rat and mouse PD exposure models. Supplementation of PD fluid with Ala-Gln resulted in reduced peritoneal thickness, αSMA expression and angiogenesis. Addition of Ala-Gln also attenuated the IL-17 pathway expression induced by PD, reflected by substantial reduction or normalization of peritoneal levels of IL-17, transforming growth factor ß, IL-6, and the transcription factor retinoic acid receptor-related orphan receptor gamma T. Moreover, increased levels of IL-17 were associated with PD-induced peritoneal thickening. Conversely, Ala-Gln treatment prevented peritoneal extracellular matrix deposition, an effect seen with IL-17 blockade. Thus, intraperitoneal administration of Ala-Gln, a stable dipeptide commonly used in parenteral nutrition, ameliorates PD-induced peritoneal damage in animal models, in part by modulating IL-17 expression. Hence, Ala-Gln supplementation of dialysate may be a potential strategy to ameliorate peritoneal deterioration during PD.


Subject(s)
Dipeptides/pharmacology , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/prevention & control , Peritoneum/drug effects , Protective Agents/pharmacology , Animals , Biomarkers/metabolism , Cytoprotection , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Interleukin-17/genetics , Male , Mice, Inbred C57BL , Neovascularization, Pathologic , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/pathology , Peritoneum/metabolism , Peritoneum/pathology , Rats, Wistar , Signal Transduction/drug effects
20.
Kidney Int ; 89(1): 21-3, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26759042

ABSTRACT

The increased awareness that disorders of phosphorus metabolism occur early in the course of chronic kidney disease fuels interest in early intervention strategies. A post hoc analysis of data from the Modification of Diet in Renal Disease (MDRD) Study questions the clinical relevance of early dietary phosphate restriction, so far considered a mainstay in the prevention and treatment of mineral metabolism disorders. Still, on the basis of available evidence, a cease-fire in the war on dietary phosphate would be premature.


Subject(s)
Phosphorus, Dietary/metabolism , Phosphorus , Diet , Humans , Kidney Failure, Chronic , Phosphates/metabolism , Renal Insufficiency, Chronic
SELECTION OF CITATIONS
SEARCH DETAIL