Pleiotropic effects of vitamin D in chronic kidney disease Wen-Chih Liu, Chia-Chao Wu, Yao-Min Hung, Min-Tser Liao, Jia-Fwu Shyu, Yuh-Feng Lin, Kuo-Cheng Lu, Kun-Chieh Yeh PII: DOI: Reference:

S0009-8981(15)30066-8 doi: 10.1016/j.cca.2015.11.029 CCA 14188

To appear in:

Clinica Chimica Acta

Received date: Revised date: Accepted date:

4 October 2015 30 November 2015 30 November 2015

Please cite this article as: Liu Wen-Chih, Wu Chia-Chao, Hung Yao-Min, Liao MinTser, Shyu Jia-Fwu, Lin Yuh-Feng, Lu Kuo-Cheng, Yeh Kun-Chieh, Pleiotropic effects of vitamin D in chronic kidney disease, Clinica Chimica Acta (2015), doi: 10.1016/j.cca.2015.11.029

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Pleiotropic effects of vitamin D in chronic kidney disease

IP

T

Authors: Wen-Chih Liua,b, Chia-Chao Wuc, Yao-Min Hungd,e, Min-Tser Liaof,g, Jia-Fwu Shyuh, Yuh-Feng Lina,i, Kuo-Cheng Luj,**,1, Kun-Chieh Yeh k,*,1

SC R

Affiliations a Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan b

MA

NU

Division of Nephrology, Department of Internal Medicine, Yonghe Cardinal Tien Hospital, New Taipei City, Taiwan c Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan d Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan Institute of Public Health, School of Medicine, National Yang Ming University, Taipei, Taiwan f Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan,

D

e

TE

Taiwan Division of Pediatrics, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan h Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan i Division of Nephrology, Department of Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan j Department of Medicine, Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic

AC

CE P

g

k

University, New Taipei City, Taiwan Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan

* Correspondence to: Dr. Kun-Chieh Yeh, Department of Surgery, Taoyuan Armed Forces General Hospital, 168, Zhongxing Road, Longtan Dist., Taoyuan City 32551, Taiwan. Tel: +886 939511532; Fax: +886 3 4799595 ** Corresponding author. E-mail addresses: [email protected] (K-C. Lu), [email protected] (K-C Yeh) 1

These authors have contributed equally to this work.

ACCEPTED MANUSCRIPT

SC R

IP

T

Abstract Low 25(OH)D levels are common in chronic kidney disease (CKD) patients and are implicated in all-cause mortality and morbidity risks. Furthermore, the progression of CKD is accompanied by a gradual decline in 25(OH)D production. Vitamin D deficiency in CKD causes skeletal disorders, such as osteoblast or osteoclast cell defects, bone turnover imbalance, and deterioration of bone quality, and nonskeletal disorders, such as metabolic syndrome, hypertension, immune dysfunction, hyperlipidemia, diabetes, and anemia.

MA

NU

Extra-renal organs possess the enzymatic machinery for converting 25(OH)D to 1,25(OH)2D, which may play considerable biological roles beyond the traditional roles of vitamin D. Pharmacological 1,25(OH)2D dose causes hypercalcemia and hyperphosphatemia as well as adynamic bone disorder, which intensifies vascular calcification. Conversely, native vitamin D supplementation reduces the risk of

TE

activity in parathyroid gland.

D

hypercalcemia and hyperphosphatemia, which may play a role in managing bone and cardio–renal health and ultimately reducing mortality in CKD patients. Nevertheless, the combination of native vitamin D and active vitamin D can enhance therapy benefits of secondary hyperparathyroidism because of extra-renal 1α-hydroxylase

AC

CE P

This article emphasizes the role of native vitamin D replacements in CKD, reviews vitamin D biology, and summarizes the present literature regarding native vitamin D replacement in the CKD population.

ACCEPTED MANUSCRIPT Introduction Chronic inflammation persists in chronic kidney disease (CKD) patients. Most CKD patients die of CKD–mineral and bone disorder (MBD) related cardiovascular events and infectious diseases. Secondary hyperparathyroidism and hypocalcemia are other common and serious complications of CKD. All of these events are likely to be associated with vitamin D deficiency. Usually, secondary hyperparathyroidism in CKD patients is caused by hypocalcemia, hyperphosphatemia, reduced 1,25(OH)2D levels, increased fibroblast growth factor-23 (FGF-23) levels, and metabolic acidosis. Vitamin D, a group of lipid-soluble compounds with a four-ringed cholesterol

SC R

IP

T

1.

MA

NU

backbone, is the only vitamin that can be synthesized by humans. Dermal synthesis and intestinal absorption are the two main vitamin D sources. Vitamin D is metabolized by 25-hydroxylase and 1-α-hydroxylase in the liver and kidneys, respectively, and converted to the active form, 1,25(OH)2D. Recent evidence suggests that the extrarenal conversion of 25(OH)D (25-hydroxyvitamin D or calcidiol) to

D

1,25(OH)2D (calcitriol ) may play considerable biological roles beyond the traditionally described roles of vitamin D [1]. Vitamin D deficiency in CKD is associated with all-cause, cardiovascular, and infectious mortality risks. Activation of the vitamin D receptor (VDR) involves several

Vitamin D metabolism and its role in CKD regulation Vitamin D deficiency is highly prevalent in CKD [2]. Therefore, clinicians

AC

2.

CE P

TE

pleiotropic effects, and then vitamin D plays new roles in maintaining health. Thus, achieving and maintaining an adequate vitamin D status may be essential in preventing cardiovascular events, balancing the bone–fat connection, correcting bone turnover disorders, improving endothelial function, alleviating proteinuria, and ultimately reducing morbidity and mortality in CKD patients.

prescribe native vitamin D supplements to CKD patients is crucial. However, overuse of active vitamin D for treating CKD affects extrarenal organs, tissues, and cells associated with vitamin D metabolism (Figure 1). 2.1 Vitamin D production and metabolism Vitamin D can be synthesized in the human skin or obtained through the diet. When the skin is adequately exposed to ultraviolet B radiation, 7-dehydrocholesterol in the skin is converted to pre-vitamin D3, which is further converted to vitamin D3 (cholecalciferol) because of body heat [3]. However, excess sunlight exposure destroys pre-vitamin D3, thus making it inactive [4]. The vitamin D binding protein (DBP) in blood delivers the skin-synthesized vitamin D3 to the liver through circulation.

ACCEPTED MANUSCRIPT

SC R

IP

T

Vitamin D from the diet can be classified into plant vitamin D2 (ergocalciferol) and animal vitamin D3 (cholecalciferol). Cholecalciferol is identical to the skin-synthesized vitamin D3. Vitamin D synthesized from the skin or obtained through the diet is reserved in fat cells [4]. Cholecalciferol and ergocalciferol are absorbed from the gastrointestinal tract, bound to chylomicrons, and delivered to the liver through hepatic portal circulation [5]. Ergocalciferol and cholecalciferol are metabolized by the enzyme vitamin D 25-hydroxylase in the liver and converted to the 25(OH)D form of 25(OH)D2 and 25(OH)D3, respectively. Clinically, 25(OH)D is the main circulating form and determines the vitamin D status in the body.

MA

NU

25(OH)D combined with DBP is delivered to the kidneys and filtrated through the glomerulus. The delivery of the 25(OH)D–DBP compound to the proximal tubular cells is facilitated by megalin receptor-mediated endocytosis [6]. Megalin is a multiligand receptor that facilitates the uptake of extracellular ligands in the renal proximal tubule. 25(OH)D is converted to its active form, calcitriol, by

D

1-α-hydroxylase and transported by the intracellular vitamin D-binding protein 3; thus, 1,25(OH)2D or 25(OH)D reenters the circulation [7, 8]. 1,25(OH)2D can induce renal megalin expression, thereby generating a cycle for conserving normal systemic 25(OH)D and 1,25(OH)2D levels [9].

AC

CE P

TE

Under normal renal function, high or low serum 25(OH)D levels do not adequately affect 1,25(OH)2D levels. However, in advanced kidney diseases (glomerular filtration rate [GFR] 30 ng/mL [12, 62-64]. Furthermore, early vitamin D supplementation can prevent or delay the onset of secondary hyperparathyroidism [65]. The KDOQI [62] and KDIGO guidelines [66] recommended that patients with low 25(OH)D and high PTH serum levels at CKD stage 3–5 should be prescribed native vitamin D. The Institute of Medicine (IOM) suggested the recommended dietary

ACCEPTED MANUSCRIPT

SC R

IP

T

allowance of vitamin D to be 600 and 800 IU/day for people aged 70 years, respectively. The therapy goal is to reach adequate 25(OH)D levels of >20 ng/mL (or 50 nmol/L) [67]. In addition, the IOM states that the upper level of intake is 4000 IU/day [68]. However, in 2010, the International Osteoporosis Foundation recommended that 25(OH)D levels >30 ng/mL (75 nmol/L) are optimal for preventing falls and fractures. The vitamin D dosage should reach 800–1,000 IU/day [69], although a higher dosage was suggested by other authors [70]. 3.3 Vitamin D effects on bone quality

MA

NU

Vitamin D deficiency is well known to lead to rickets in children and osteomalacia or osteoporosis in adults. Previous studies approve that vitamin D supplement cures weakened bone mineralization. Osteoporosis is a systemic metabolic bone disease of multiple causes, which have the characters of impaired bone strength and increasing fragility fracture risk. Bone strength is reflected equally

D

by both BMD, which is determined by peak bone mass and amount of bone loss and bone quality, which refers to architecture, turnover, and mineralization [71, 72]. Mounting evidence indicates most bone cells possess 1α-hydroxylase to perform the intracrine/paracrine activities of vitamin D [73]. 1,25(OH)2D produced by

AC

CE P

TE

bone cells can diminish bone resorption through an intracrine/paracrine function resulting in increased bone mass [74]. Vitamin D deficiency will increase the risks of fracture, which not only impairs bone mineralization but also numerous pathological alterations. Vitamin D deficiency increases both the beginning and extension of fragility fracture. Thus, vitamin D deficiency is associated with both diminished bone mass and worsen bone quality. Appropriate vitamin D levels are the key to preserve the structural integrity of bone [75]. Although several meta-analyses show no influence of vitamin D treatment alone on fracture risk [76, 77], the emerging evidence shows that cholecalciferol combined with calcium has the greatest advantage to reduce the risk of nonvertebral and hip fractures [78]. 4. Nonskeletal effects of vitamin D 4.1 Vitamin D in bone–fat connection Low 25(OH)D3 levels are associated with metabolic syndromes, such as overweight, hypertension, high blood glucose, and hyperlipidemia [79]. Obesity exacerbates CKD and osteoporosis, and these two disorders frequently induce severe bone loss. Bone loss is more severe in CKD patients with vitamin D deficiency than in patients without vitamin D deficiency [80] (Figure 3). Adipocytes are formed from MSCs, which can differentiate into different cells,

ACCEPTED MANUSCRIPT

SC R

IP

T

such as osteoblasts, chondrocytes, and myocytes [81] (Figure 3). MSCs differentiate in a complicated manner into osteoblasts or adipocytes and are influenced by several growth factors, C/EBP-δ, C/EBP-α, and peroxisome proliferator-activated receptor-γ (PPAR-γ) in the bone marrow. Adipogenesis and osteoblastogenesis have an inverse association and are delicately balanced [82]. Obesity and aging are clearly associated with greater adipogenesis potential and less osteoblastogenesis from MSCs in the bone marrow [83, 84]. In elderly people with severe bone loss conditions such as osteoporosis, MSCs tend to undergo adipogenesis in the bone marrow [83, 84]. Noggin, a glycosylated protein that inhibits bone morphogenetic protein signaling,

MA

NU

directly induces adipogenesis from MSCs. Thus, high noggin levels may be used as a potential therapeutic target, and noggin may serve as a potential substitute biomarker in the obese population [85]. By contrast, the Wnt signaling pathway promotes osteogenesis and inhibits adipogenesis synchronously [86, 87]. Furthermore, these two pathways can influence

D

each other. The canonical Wnt signaling in bone can prevent the phosphorylation of β-catenin and its proteosomal degradation, and several reports have consistently stated that the Wnt/β-catenin activity is essential for bone development [88]. In addition, Wnt10b is crucial to bone formation because it inhibits PPAR-γ expression,

CE P

TE

enhances osteoblastogenesis, and increases the bone density in the bone marrow [89, 90]. Therefore, use of vitamin D supplements improves osteoblast differentiation and function and induces adipocyte apoptosis, resulting in increased bone mass in the bone marrow and reducing the likelihood of obesity development.

AC

4.2 Vitamin D and blood pressure Low vitamin D levels have been associated with increased vascular stiffness, endothelial dysfunction, inflammatory cytokines, and higher coronary artery calcium scores [91]. In a previous study on patients with hypertension exposed to sufficient sunlight, the 25(OH)D levels were upregulated by approximately 180%, and subsequently the blood pressure was normal [92]. In an animal experiment, VDR-knockout mice had enhanced renin-angiotensin-aldosterone system (RAAS) signaling in the blood, which led to significant sodium retention, vascular resistance, and hypertension [93]. 1,25(OH)2D produced from the kidney enters the circulation and reduces renin production in the kidneys to control the blood pressure [4]. Another study reported that weekly native vitamin D supplementation (cholecalciferol; 50,000 units for 12 weeks) in type 2 diabetic patients may improve hypertension [94]. 4.3 Vitamin D and immune function

ACCEPTED MANUSCRIPT

SC R

IP

T

More and more researches state that vitamin D deficiency not only causes dysregulation of the innate and adaptive immune systems, but also promotes microinflammation in CKD [95]. Owing to immune cells carrying VDR and 1α-hydroxylase, which can produce the active metabolite 1,25(OH)2D through local synthesis. In innate immunity, vitamin D promotes macrophages to produce cathelicidin and β-defensin 2 and enhance the capacity for autophagy via toll-like receptor activation as well as affects complement concentrations [96]. Therefore, 1,25(OH)2D was proved to heighten the effects of macrophages and monocytes against pathogens by stimulating monocytes change into mature phagocytic

MA

NU

macrophages [97]. In addition, in adaptive immunity, vitamin D suppresses the maturation of dendritic cells (DCs) and weakens antigen presentation. Vitamin D will increase T helper (Th) 2 cytokine production and the efficiency of Treg lymphocytes [98] but diminish the secretion of Th1 and Th17 cytokines to decrease autoimmune disease [99].

D

Interestingly, vitamin D plays a role in the crosstalk between innate and adaptive immunity because it has a significant impact on macrophages and DCs [96]. DCs are the important connector between innate and adaptive immune systems. Local production of 1,25(OH)2D in paracrine responses also seems to have an effect on

CE P

TE

monocytes and immature DCs. Thus, local 1,25(OH)2D is assumed to enhance a tolerogenic immune response and possess immunosuppressive properties.

AC

4.4 Vitamin D and diabetes mellitus nephropathy Because of secondary hyperparathyroidism, inflammation, and oxidative stress, CKD patients have impaired insulin secretion and enhanced insulin resistance, which alter glucose metabolism [100]. Active vitamin D (1,25(OH)2D) directly enhances insulin secretion by interacting with the 1,25(OH)2D3-RXR-VDR complex, which binds to vitamin D responsive elements, thus enhancing the transcriptional activation of the insulin gene, increasing insulin synthesis, and indirectly increasing the calcium concentration within the β islet cells of the pancreas. In addition, native vitamin D augments insulin sensitivity by stimulating the insulin receptor and activating PPAR-γ [101]. In a previous study on people with prediabetes, vitamin D and calcium upregulated insulin sensitivity [102]. 4.5 Vitamin D in the endothelium The vascular endothelial function of CKD patients is dysregulated. Active vitamin D inhibits vascular calcification through two possible mechanisms. First, increased Klotho secretion can alleviate phosphaturia and restrict phosphate uptake by the vascular smooth muscles. Second, upregulating the expression of the calcification

ACCEPTED MANUSCRIPT

SC R

IP

T

inhibitor osteopontin in aortic medial cells and increasing matrix G1A protein synthesis can improve vascular endothelial function [103, 104]. Chitalia et al. reported that oral vitamin D (cholecalciferol) improves endothelial vasomotor and secretory functions in CKD patients without significant adverse effects on the arterial stiffness [105]. In addition, vitamin D3 administration (cholecalciferol, 4000 IU/day) enhanced vascular regeneration by inducing stromal cell–derived factor 1 expression in the healthy population. Therefore, native vitamin D3 is a new approach for promoting vascular endothelial repair [106].

MA

NU

4.6 Vitamin D and albuminuria As CKD progresses, albuminuria, including microalbuminuria, increases [107, 108]. Therefore, albuminuria is a critical indicator of kidney deterioration and subsequent cardiovascular diseases, thus serving as a potential treatment target [109]. Traditionally, using RAAS blockers, reducing salt intake, and controlling blood

D

pressure can reduce albuminuria. However, the effects of these methods on preventing kidney damage are limited. Vitamin D deficiency is related to albuminuria and CKD progression [35, 110-112]. Furthermore, vitamin D deficiency independently predicts the 5-year

AC

CE P

TE

incidence of albuminuria [113]. Activation of the VDR is essential in reducing proteinuria [114, 115]. Molina et al. reported that vitamin D supplements may effectively reduce albuminuria at CKD stages 3–4 [116]. Massart et al. reported that oral administration of 25,000 IU/week of cholecalciferol for 13 weeks increases 25(OH)D and 1,25(OH)2D levels in patients on hemodialysis [43]. Hence, vitamin D supplementation is indicated in CKD. 4.7 Vitamin D and anemia Chronic inflammation and a reduced GFR in CKD patients cause iron imbalance and increased hepcidin production [117] (Figure 4). Vitamin D deficiency alters innate and adaptive immune function, which will increase inflammatory cytokine production (IL-6, IFN-γ, TFN-α) which will stimulate canonical janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, and phosphorylation of JAKs and STAT3 to stimulate hepcidin expression in liver [118, 119] . Hepcidin limits iron absorption from the intestine or iron release from macrophages and liver reserves by inducing ferroportin degradation for maintaining the systemic iron balance [120]. Under iron deficiency, anemia or hypoxia prohibits hepcidin production, increasing iron usability in the liver; however, inflammation and the use of excess iron supplements stimulate hepcidin production, thus inhibiting ferroportin activity and limiting iron usability [121]. Several factors, such as the use of

ACCEPTED MANUSCRIPT

SC R

IP

T

phosphate binders and antacids, loss of blood during hemodialysis, and intake of erythropoiesis-stimulating agents (ESA), cause iron deficiency [122]. In addition, secondary hyperparathyroidism will directly inhibit erythroid progenitors, endogenous EPO synthesis, and RBC survival [123]. It also indirectly promotes bone marrow fibrosis, hyperphosphataemia, and increase serum alkaline phosphatase. All of them will lead to ESA hyporesponsivenes [124]. According to recent studies, vitamin D deficiency, low hemoglobin levels, and ESA resistance constitute a pathophysiological cofactor of renal anemia [125, 126]. Providing vitamin D or active vitamin D is associated with improving anemia and

MA

NU

reducing ESA requirements [127, 128]. Therefore, vitamin D levels and ESA requirements exhibit an inverse relationship in CKD patients [129]. In addition, vitamin D application promotes anti-inflammation and erythroid proliferation [13, 130].

D

4.8 Vitamin D and lipid metabolism The VDR affects bile acid synthesis and reduces cholesterol levels in hepatocytes and serum. Activation of the VDR by 1,25(OH)2D reduces liver and serum cholesterol because it suppresses the expression of small heterodimer partner (SHP) and the

AC

CE P

TE

activation of cholesterol 7-α-hydroxylase (CYP7A1) [131]. SHP combines with the liver receptor homolog-1, a positive regulator of CYP7A1, for to blocking the activation of the CYP7A1 gene [132]. CYP7A1 is the rate-limiting enzyme in bile acid synthesis, and its expression controls serum cholesterol levels [133, 134]. In addition, the hepatic farnesoid X receptor (FXR) can enhance SHP activity to inhibit CYP7A1 [135, 136]. A study showed that VDR activation downregulated FXR and SHP expression [135], which are responsible for increasing CYP7A1 expression to lower cholesterol. 5. Combination of native and active vitamin D therapy in CKD 25(OH)D deficiency is widespread in the general and CKD populations [137]. Native vitamin D supplementation prevents secondary hyperparathyroidism in early CKD; KDOQI suggests its use is not beneficial in advanced CKD because of the lack of 1-α-hydroxylase in the kidneys [62, 138]. However, another recent study indicated that 1,25(OH)2D levels were increased after supplementation with native vitamin D in hemodialysis patients and suggested that there was enough extra-renal 1-α-hydroxylase activity to produce serum levels of 1,25(OH)2D even in ESRD [139]. Physicians should prescribe native vitamin D (cholecalciferol, ergocalciferol, or calcidiol) to all patients whose 25(OH)D levels are 400,000 IU and loading doses ≤ 300 000 IU should be advocated [150]. The symptoms of hypercalcemia induced by vitamin D intoxication include confusion, polyuria, polydipsia, anorexia, vomiting, and muscle weakness. The hypercalcemia of long term vitamin D intoxication may cause nephrocalcinosis, bone demineralization, pain, and [151]. In addition, vitamin D intoxication can induce acute renal failure [152]. 7.

Conclusion Several studies have recently reported the advantages of vitamin D in treating CKD-MBD. Vitamin D, a hormone with pleiotropic functions, is a critical substance that provides nourishment essential for bone health and growth. Native vitamin D use has fewer hypercalcemia- and hyperphosphatemia-associated complications than active vitamin D use does. In addition, vitamin D exerts beneficial quality on

ACCEPTED MANUSCRIPT

SC R

IP

T

bone cells and enhances osteoblast cell production, thereby alleviating osteoporosis. Reducing adipogenesis, alleviating metabolic syndrome, maintaining regular blood pressure, improving insulin resistance and alleviating diabetes mellitus, lowering cholesterol levels, increasing vascular endothelial cell function, and managing renal anemia are other functions of vitamin D. For early-stage CKD patients, adequate native vitamin D supplementation decelerates the decline of kidney function by slowing the progression to the ESRD stage, relieves proteinuria caused by CKD, and reduces the occurrence of hyperparathyroidism. In advance-stage CKD patients with high bone turnover, a

MA

NU

combination therapy of native vitamin D and active vitamin D can reduce the dosage of active vitamin D and vascular calcification. In addition, native vitamin D supplementation will enhance the function of osteoblasts and improve the bone quality, and increase the viability of osteoblasts in patients with low bone turnover. In summary, native vitamin D promotes bone remodeling and enhances the health of

D

nonskeletal organs. Therefore, its role is extremely valuable for bone metabolism and maintaining bone quality in CKD. Conflict of interests

AC

CE P

TE

The authors declare that there is no conflict of interests regarding the publication of this paper.

ACCEPTED MANUSCRIPT

9.

10.

11. 12. 13. 14. 15.

16.

NU

MA

D

8.

D. The American journal of clinical nutrition 2004; 80: 1689S-96S. Dusso AS, Tokumoto M: Defective renal maintenance of the vitamin D endocrine system impairs vitamin D renoprotection: a downward spiral in kidney disease. Kidney international 2011; 79: 715-29. Boros S, Bindels RJ, Hoenderop JG: Active Ca(2+) reabsorption in the connecting tubule. Pflugers Archiv : European journal of physiology 2009; 458: 99-109. Deluca HF: History of the discovery of vitamin D and its active metabolites.

TE

7.

507-15. Holick MF: Resurrection of vitamin D deficiency and rickets. The Journal of clinical investigation 2006; 116: 2062-72. DeLuca HF: Overview of general physiologic features and functions of vitamin

CE P

6.

357: 266-81. Bhan I, Hewison M, Thadhani R: Dietary vitamin D intake in advanced CKD/ESRD. Seminars in dialysis 2010; 23: 407-10. Nykjaer A, Dragun D, Walther D et al: An endocytic pathway essential for renal uptake and activation of the steroid 25-(OH) vitamin D3. Cell 1999; 96:

AC

5.

SC R

IP

T

References: 1. Galassi A, Bellasi A, Auricchio S, Papagni S, Cozzolino M: Which vitamin D in CKD-MBD? The time of burning questions. BioMed research international 2013; 2013: 864012. 2. Zhu N, Wang J, Gu L, Wang L, Yuan W: Vitamin D supplements in chronic kidney disease. Renal failure 2015: 1-8. 3. Holick MF: Vitamin D: a D-Lightful health perspective. Nutrition reviews 2008; 66: S182-94. 4. Holick MF: Vitamin D deficiency. The New England journal of medicine 2007;

BoneKEy reports 2014; 3: 479. Dusso AS, Brown AJ, Slatopolsky E: Vitamin D. American journal of physiology Renal physiology 2005; 289: F8-28. Andress DL: Vitamin D in chronic kidney disease: a systemic role for selective vitamin D receptor activation. Kidney international 2006; 69: 33-43. Liu PT, Stenger S, Li H et al: Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science 2006; 311: 1770-3. Hewison M, Burke F, Evans KN et al: Extra-renal 25-hydroxyvitamin D3-1alpha-hydroxylase in human health and disease. The Journal of steroid biochemistry and molecular biology 2007; 103: 316-21. Rojas-Rivera J, De La Piedra C, Ramos A, Ortiz A, Egido J: The expanding spectrum of biological actions of vitamin D. Nephrology, dialysis,

ACCEPTED MANUSCRIPT

20.

21. 22.

T

IP

Kalantar-Zadeh K, Kovesdy CP: Clinical outcomes with active versus nutritional vitamin D compounds in chronic kidney disease. Clinical journal of the American Society of Nephrology : CJASN 2009; 4: 1529-39. Holick MF: Vitamin D: extraskeletal health. Rheumatic diseases clinics of North America 2012; 38: 141-60. Harms LR, Burne TH, Eyles DW, McGrath JJ: Vitamin D and the brain. Best practice & research Clinical endocrinology & metabolism 2011; 25: 657-69. Girgis CM, Clifton-Bligh RJ, Turner N, Lau SL, Gunton JE: Effects of vitamin D in skeletal muscle: falls, strength, athletic performance and insulin sensitivity.

D

23.

SC R

19.

NU

18.

MA

17.

transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association 2010; 25: 2850-65. Nigwekar SU, Bhan I, Thadhani R: Ergocalciferol and cholecalciferol in CKD. American journal of kidney diseases : the official journal of the National Kidney Foundation 2012; 60: 139-56. Hossein-nezhad A, Holick MF: Vitamin D for health: a global perspective. Mayo Clinic proceedings 2013; 88: 720-55. Gunta SS, Thadhani RI, Mak RH: The effect of vitamin D status on risk factors for cardiovascular disease. Nature reviews Nephrology 2013; 9: 337-47.

26. 27.

28. 29. 30.

31.

TE

CE P

25.

AC

24.

Clinical endocrinology 2014; 80: 169-81. Norman PE, Powell JT: Vitamin D and cardiovascular disease. Circulation research 2014; 114: 379-93. Li YC: Vitamin D regulation of the renin-angiotensin system. Journal of cellular biochemistry 2003; 88: 327-31. Zittermann A: Vitamin D in preventive medicine: are we ignoring the evidence? The British journal of nutrition 2003; 89: 552-72. Martineau AR, Honecker FU, Wilkinson RJ, Griffiths CJ: Vitamin D in the treatment of pulmonary tuberculosis. The Journal of steroid biochemistry and molecular biology 2007; 103: 793-8. Brown AJ, Dusso A, Slatopolsky E: Vitamin D. The American journal of physiology 1999; 277: F157-75. Aranda A, Pascual A: Nuclear hormone receptors and gene expression. Physiological reviews 2001; 81: 1269-304. Burke LJ, Baniahmad A: Co-repressors 2000. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 2000; 14: 1876-88. Carlberg C, Campbell MJ: Vitamin D receptor signaling mechanisms: integrated actions of a well-defined transcription factor. Steroids 2013; 78: 127-36.

ACCEPTED MANUSCRIPT

34.

T

38.

abnormalities of mineral homeostasis, vitamin D and parathyroid hormone in a cohort of pre-dialysis patients. The chronic renal impairment in Birmingham (CRIB) study. Nephron Clinical practice 2007; 107: c109-16. Christakos S, Ajibade DV, Dhawan P, Fechner AJ, Mady LJ: Vitamin D:

40.

41.

42.

43.

MA

D

TE

39.

metabolism. Endocrinology and metabolism clinics of North America 2010; 39: 243-53, table of contents. Zierold C, Darwish HM, DeLuca HF: Identification of a vitamin D-response element in the rat calcidiol (25-hydroxyvitamin D3) 24-hydroxylase gene. Proceedings of the National Academy of Sciences of the United States of America 1994; 91: 900-2. Tourigny A, Charbonneau F, Xing P et al: CYP24A1 exacerbated activity during diabetes contributes to kidney tubular apoptosis via caspase-3 increased

CE P

36.

NU

37.

Ravani P, Malberti F, Tripepi G et al: Vitamin D levels and patient outcome in chronic kidney disease. Kidney international 2009; 75: 88-95. Wolf M, Shah A, Gutierrez O et al: Vitamin D levels and early mortality among incident hemodialysis patients. Kidney international 2007; 72: 1004-13. Zehnder D, Landray MJ, Wheeler DC et al: Cross-sectional analysis of

AC

35.

IP

33.

Chronic Kidney Disease Prognosis C, Matsushita K, van der Velde M et al: Association of estimated glomerular filtration rate and albuminuria with all-cause and cardiovascular mortality in general population cohorts: a collaborative meta-analysis. Lancet 2010; 375: 2073-81. Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY: Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. The New England journal of medicine 2004; 351: 1296-305. Nigwekar SU, Tamez H, Thadhani RI: Vitamin D and chronic kidney disease-mineral bone disease (CKD-MBD). BoneKEy reports 2014; 3: 498.

SC R

32.

expression and activation. PloS one 2012; 7: e48652. Larsson T, Nisbeth U, Ljunggren O, Juppner H, Jonsson KB: Circulating concentration of FGF-23 increases as renal function declines in patients with chronic kidney disease, but does not change in response to variation in phosphate intake in healthy volunteers. Kidney international 2003; 64: 2272-9. Prie D, Friedlander G: Reciprocal control of 1,25-dihydroxyvitamin D and FGF23 formation involving the FGF23/Klotho system. Clinical journal of the American Society of Nephrology : CJASN 2010; 5: 1717-22. Massart A, Debelle FD, Racape J et al: Biochemical parameters after cholecalciferol repletion in hemodialysis: results From the VitaDial randomized trial. American journal of kidney diseases : the official journal of

ACCEPTED MANUSCRIPT

44.

osteoblast-like cells. Calcified tissue international 1991; 49: 35-42. Reinhardt TA, Horst RL: Parathyroid hormone down-regulates 1,25-dihydroxyvitamin D receptors (VDR) and VDR messenger ribonucleic acid in vitro and blocks homologous up-regulation of VDR in vivo. Endocrinology 1990; 127: 942-8.

MA

NU

47.

SC R

46.

IP

T

45.

the National Kidney Foundation 2014; 64: 696-705. Ding C, Gao D, Wilding J, Trayhurn P, Bing C: Vitamin D signalling in adipose tissue. The British journal of nutrition 2012; 108: 1915-23. Ishimura E, Nishizawa Y, Inaba M et al: Serum levels of 1,25-dihydroxyvitamin D, 24,25-dihydroxyvitamin D, and 25-hydroxyvitamin D in nondialyzed patients with chronic renal failure. Kidney international 1999; 55: 1019-27. van Leeuwen JP, Pols HA, Schilte JP, Visser TJ, Birkenhager JC: Modulation by epidermal growth factor of the basal 1,25(OH)2D3 receptor level and the heterologous up-regulation of the 1,25(OH)2D3 receptor in clonal

Godschalk M, Levy JR, Downs RW, Jr.: Glucocorticoids decrease vitamin D receptor number and gene expression in human osteosarcoma cells. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research 1992; 7: 21-7.

49.

van Driel M, Koedam M, Buurman CJ et al: Evidence for auto/paracrine actions of vitamin D in bone: 1alpha-hydroxylase expression and activity in human bone cells. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 2006; 20: 2417-9. Haussler MR, Haussler CA, Whitfield GK et al: The nuclear vitamin D receptor controls the expression of genes encoding factors which feed the "Fountain of Youth" to mediate healthful aging. The Journal of steroid biochemistry and molecular biology 2010; 121: 88-97.

51.

52.

53.

TE

CE P

AC

50.

D

48.

Fretz JA, Zella LA, Kim S, Shevde NK, Pike JW: 1,25-Dihydroxyvitamin D3 induces expression of the Wnt signaling co-regulator LRP5 via regulatory elements located significantly downstream of the gene's transcriptional start site. The Journal of steroid biochemistry and molecular biology 2007; 103: 440-5. Paredes R, Arriagada G, Cruzat F et al: The Runx2 transcription factor plays a key role in the 1alpha,25-dihydroxy Vitamin D3-dependent upregulation of the rat osteocalcin (OC) gene expression in osteoblastic cells. The Journal of steroid biochemistry and molecular biology 2004; 89-90: 269-71. Shen Q, Christakos S: The vitamin D receptor, Runx2, and the Notch signaling pathway cooperate in the transcriptional regulation of osteopontin. The Journal of biological chemistry 2005; 280: 40589-98.

ACCEPTED MANUSCRIPT

up-regulation of its receptor in rat osteosarcoma cells. Biochimica et biophysica acta 1988; 970: 122-9. Staal A, van den Bemd GJ, Birkenhager JC, Pols HA, van Leeuwen JP: Consequences of vitamin D receptor regulation for the 1,25-dihydroxyvitamin

D

58.

NU

57.

action of 1,25-dihydroxyvitamin D(3) in human marrow stromal cells. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research 2012; 27: 1992-2000. Pols HA, Birkenhager JC, Schilte JP, Visser TJ: Evidence that the self-induced metabolism of 1,25-dihydroxyvitamin D-3 limits the homologous

MA

56.

SC R

IP

55.

van Driel M, Koedam M, Buurman CJ et al: Evidence that both 1alpha,25-dihydroxyvitamin D3 and 24-hydroxylated D3 enhance human osteoblast differentiation and mineralization. Journal of cellular biochemistry 2006; 99: 922-35. Zhou YS, Liu YS, Tan JG: Is 1, 25-dihydroxyvitamin D3 an ideal substitute for dexamethasone for inducing osteogenic differentiation of human adipose tissue-derived stromal cells in vitro? Chinese medical journal 2006; 119: 1278-86. Zhou S, Glowacki J, Kim SW et al: Clinical characteristics influence in vitro

T

54.

61.

62.

63.

64. 65.

TE

CE P

60.

AC

59.

D3-induced 24-hydroxylase activity in osteoblast-like cells: initiation of the C24-oxidation pathway. Bone 1997; 20: 237-43. Takeda S, Yoshizawa T, Nagai Y et al: Stimulation of osteoclast formation by 1,25-dihydroxyvitamin D requires its binding to vitamin D receptor (VDR) in osteoblastic cells: studies using VDR knockout mice. Endocrinology 1999; 140: 1005-8. Helvig CF, Cuerrier D, Hosfield CM et al: Dysregulation of renal vitamin D metabolism in the uremic rat. Kidney international 2010; 78: 463-72. Suda T, Takahashi F, Takahashi N: Bone effects of vitamin D - Discrepancies between in vivo and in vitro studies. Archives of biochemistry and biophysics 2012; 523: 22-9. National Kidney F: K/DOQI clinical practice guidelines for bone metabolism and disease in chronic kidney disease. American journal of kidney diseases : the official journal of the National Kidney Foundation 2003; 42: S1-201. Brown AJ: Therapeutic uses of vitamin D analogues. American journal of kidney diseases : the official journal of the National Kidney Foundation 2001; 38: S3-S19. Holick MF: Vitamin D for health and in chronic kidney disease. Seminars in dialysis 2005; 18: 266-75. Shroff R, Wan M, Gullett A et al: Ergocalciferol supplementation in children

ACCEPTED MANUSCRIPT

SC R

Dawson-Hughes B, Mithal A, Bonjour JP et al: IOF position statement: vitamin D recommendations for older adults. Osteoporosis international : a journal established as result of cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA 2010; 21:

D

69.

NU

68.

1,25(OH)2D3 administration alone. Mineral and electrolyte metabolism 1983; 9: 19-27. Mazzaferro S, Goldsmith D, Larsson TE, Massy ZA, Cozzolino M: Vitamin D metabolites and/or analogs: which D for which patient? Current vascular pharmacology 2014; 12: 339-49.

MA

67.

IP

T

66.

with CKD delays the onset of secondary hyperparathyroidism: a randomized trial. Clinical journal of the American Society of Nephrology : CJASN 2012; 7: 216-23. Kidney Disease: Improving Global Outcomes CKDMBDWG: KDIGO clinical practice guideline for the diagnosis, evaluation, prevention, and treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD). Kidney international Supplement 2009: S1-130. Coen G, Taccone Gallucci M, Bonucci E et al: 1,25(OH)2D3 and 25-OHD3 in the treatment of renal osteodystrophy: comparison of combined versus

72.

73.

74.

75.

TE

CE P

71.

AC

70.

1151-4. Tangpricha V, Wasse H: Vitamin D therapy in kidney disease: more vitamin D is necessary. American journal of kidney diseases : the official journal of the National Kidney Foundation 2014; 64: 667-9. Miller PD, Bolognese MA, Lewiecki EM et al: Effect of denosumab on bone density and turnover in postmenopausal women with low bone mass after long-term continued, discontinued, and restarting of therapy: a randomized blinded phase 2 clinical trial. Bone 2008; 43: 222-9.

Miller PD: Bone disease in CKD: a focus on osteoporosis diagnosis and management. American journal of kidney diseases : the official journal of the National Kidney Foundation 2014; 64: 290-304. Anderson PH, Lam NN, Turner AG et al: The pleiotropic effects of vitamin D in bone. The Journal of steroid biochemistry and molecular biology 2013; 136: 190-4. Glendenning P, Inderjeeth CA: Controversy and consensus regarding vitamin D: Recent methodological changes and the risks and benefits of vitamin D supplementation. Critical reviews in clinical laboratory sciences 2015: 1-16. Busse B, Bale HA, Zimmermann EA et al: Vitamin D deficiency induces early signs of aging in human bone, increasing the risk of fracture. Science translational medicine 2013; 5: 193ra88.

ACCEPTED MANUSCRIPT 76.

82.

83.

SC R

NU

Takada I, Kouzmenko AP, Kato S: Molecular switching of osteoblastogenesis versus adipogenesis: implications for targeted therapies. Expert opinion on therapeutic targets 2009; 13: 593-603. Muruganandan S, Roman AA, Sinal CJ: Adipocyte differentiation of bone marrow-derived mesenchymal stem cells: cross talk with the osteoblastogenic program. Cellular and molecular life sciences : CMLS 2009; 66: 236-53. Wu XB, Li Y, Schneider A et al: Impaired osteoblastic differentiation, reduced bone formation, and severe osteoporosis in noggin-overexpressing mice. The Journal of clinical investigation 2003; 112: 924-34. Kajkenova O, Lecka-Czernik B, Gubrij I et al: Increased adipogenesis and myelopoiesis in the bone marrow of SAMP6, a murine model of defective osteoblastogenesis and low turnover osteopenia. Journal of bone and mineral

AC

84.

MA

81.

D

80.

TE

79.

2007; 370: 657-66. Challoumas D: Vitamin D supplementation and lipid profile: what does the best available evidence show? Atherosclerosis 2014; 235: 130-9. Bouillon R, Carmeliet G, Lieben L et al: Vitamin D and energy homeostasis: of mice and men. Nature reviews Endocrinology 2014; 10: 79-87.

CE P

78.

IP

T

77.

Theodoratou E, Tzoulaki I, Zgaga L, Ioannidis JP: Vitamin D and multiple health outcomes: umbrella review of systematic reviews and meta-analyses of observational studies and randomised trials. Bmj 2014; 348: g2035. Bolland MJ, Grey A, Gamble GD, Reid IR: The effect of vitamin D supplementation on skeletal, vascular, or cancer outcomes: a trial sequential meta-analysis. The lancet Diabetes & endocrinology 2014; 2: 307-20. Tang BM, Eslick GD, Nowson C, Smith C, Bensoussan A: Use of calcium or calcium in combination with vitamin D supplementation to prevent fractures and bone loss in people aged 50 years and older: a meta-analysis. Lancet

85.

86. 87. 88.

research : the official journal of the American Society for Bone and Mineral Research 1997; 12: 1772-9. Sawant A, Chanda D, Isayeva T et al: Noggin is novel inducer of mesenchymal stem cell adipogenesis: implications for bone health and obesity. The Journal of biological chemistry 2012; 287: 12241-9. Bennett CN, Ross SE, Longo KA et al: Regulation of Wnt signaling during adipogenesis. The Journal of biological chemistry 2002; 277: 30998-1004. Gimble JM, Zvonic S, Floyd ZE, Kassem M, Nuttall ME: Playing with bone and fat. Journal of cellular biochemistry 2006; 98: 251-66. Day TF, Guo X, Garrett-Beal L, Yang Y: Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Developmental cell 2005; 8: 739-50.

ACCEPTED MANUSCRIPT

92. 93.

T

IP

2014; 167: 283-91. Krause R, Buhring M, Hopfenmuller W, Holick MF, Sharma AM: Ultraviolet B and blood pressure. Lancet 1998; 352: 709-10. Li YC, Kong J, Wei M et al: 1,25-Dihydroxyvitamin D(3) is a negative endocrine regulator of the renin-angiotensin system. The Journal of clinical investigation 2002; 110: 229-38. Nasri H, Behradmanesh S, Ahmadi A, Rafieian-Kopaei M: Impact of oral vitamin D (cholecalciferol) replacement therapy on blood pressure in type 2 diabetes patients; a randomized, double-blind, placebo controlled clinical trial.

D

94.

SC R

91.

NU

90.

Kang S, Bennett CN, Gerin I et al: Wnt signaling stimulates osteoblastogenesis of mesenchymal precursors by suppressing CCAAT/enhancer-binding protein alpha and peroxisome proliferator-activated receptor gamma. The Journal of biological chemistry 2007; 282: 14515-24. Andrade AC, Nilsson O, Barnes KM, Baron J: Wnt gene expression in the post-natal growth plate: regulation with chondrocyte differentiation. Bone 2007; 40: 1361-9. Kunadian V, Ford GA, Bawamia B, Qiu W, Manson JE: Vitamin D deficiency and coronary artery disease: a review of the evidence. American heart journal

MA

89.

97.

98. 99.

100. 101.

TE

CE P

96.

AC

95.

Journal of nephropathology 2014; 3: 29-33. Peterson CA, Heffernan ME: Serum tumor necrosis factor-alpha concentrations are negatively correlated with serum 25(OH)D concentrations in healthy women. Journal of inflammation 2008; 5: 10. Liu WC, Zheng CM, Lu CL et al: Vitamin D and immune function in chronic kidney disease. Clinica chimica acta; international journal of clinical chemistry 2015; 450: 135-44. Koeffler HP, Amatruda T, Ikekawa N, Kobayashi Y, DeLuca HF: Induction of macrophage differentiation of human normal and leukemic myeloid stem cells by 1,25-dihydroxyvitamin D3 and its fluorinated analogues. Cancer research 1984; 44: 5624-8. Hewison M: Vitamin D and innate and adaptive immunity. Vitamins and hormones 2011; 86: 23-62. Antico A, Tampoia M, Tozzoli R, Bizzaro N: Can supplementation with vitamin D reduce the risk or modify the course of autoimmune diseases? A systematic review of the literature. Autoimmunity reviews 2012; 12: 127-36. Nigwekar SU, Thadhani RI: Shining light on vitamin D trials in chronic kidney disease. Kidney international 2013; 83: 198-200. Harinarayan CV: Vitamin D and diabetes mellitus. Hormones 2014; 13: 163-81.

ACCEPTED MANUSCRIPT

105.

106.

T

IP

level. Cytokine 1992; 4: 506-12. Chitalia N, Ismail T, Tooth L et al: Impact of vitamin D supplementation on arterial vasomotion, stiffness and endothelial biomarkers in chronic kidney disease patients. PloS one 2014; 9: e91363. Wong MS, Leisegang MS, Kruse C et al: Vitamin D promotes vascular regeneration. Circulation 2014; 130: 976-86. Astor BC, Matsushita K, Gansevoort RT et al: Lower estimated glomerular filtration rate and higher albuminuria are associated with mortality and end-stage renal disease. A collaborative meta-analysis of kidney disease

D

107.

SC R

104.

NU

103.

Gagnon C, Daly RM, Carpentier A et al: Effects of combined calcium and vitamin D supplementation on insulin secretion, insulin sensitivity and beta-cell function in multi-ethnic vitamin D-deficient adults at risk for type 2 diabetes: a pilot randomized, placebo-controlled trial. PloS one 2014; 9: e109607. Komaba H, Fukagawa M: Vitamin D and secreted Klotho: a long-awaited panacea for vascular calcification? Kidney international 2012; 82: 1248-50. Muller K, Haahr PM, Diamant M et al: 1,25-Dihydroxyvitamin D3 inhibits cytokine production by human blood monocytes at the post-transcriptional

MA

102.

110.

111.

112.

113.

TE

CE P

109.

AC

108.

population cohorts. Kidney international 2011; 79: 1331-40. Hemmelgarn BR, Manns BJ, Lloyd A et al: Relation between kidney function, proteinuria, and adverse outcomes. Jama 2010; 303: 423-9. de Zeeuw D, Remuzzi G, Parving HH et al: Proteinuria, a target for renoprotection in patients with type 2 diabetic nephropathy: lessons from RENAAL. Kidney international 2004; 65: 2309-20. Gorriz JL, Molina P, Bover J et al: Characteristics of bone mineral metabolism in patients with stage 3-5 chronic kidney disease not on dialysis: results of the OSERCE study. Nefrologia : publicacion oficial de la Sociedad Espanola Nefrologia 2013; 33: 46-60. Melamed ML, Astor B, Michos ED et al: 25-hydroxyvitamin D levels, race, and the progression of kidney disease. Journal of the American Society of Nephrology : JASN 2009; 20: 2631-9. Holden RM, Morton AR, Garland JS et al: Vitamins K and D status in stages 3-5 chronic kidney disease. Clinical journal of the American Society of Nephrology : CJASN 2010; 5: 590-7. Damasiewicz MJ, Magliano DJ, Daly RM et al: Serum 25-hydroxyvitamin D deficiency and the 5-year incidence of CKD. American journal of kidney diseases : the official journal of the National Kidney Foundation 2013; 62: 58-66.

ACCEPTED MANUSCRIPT

116.

de Zeeuw D, Agarwal R, Amdahl M et al: Selective vitamin D receptor activation with paricalcitol for reduction of albuminuria in patients with type 2 diabetes (VITAL study): a randomised controlled trial. Lancet 2010; 376: 1543-51. Fishbane S, Chittineni H, Packman M et al: Oral paricalcitol in the treatment of patients with CKD and proteinuria: a randomized trial. American journal of kidney diseases : the official journal of the National Kidney Foundation 2009; 54: 647-52. Molina P, Gorriz JL, Molina MD et al: The effect of cholecalciferol for lowering

117.

albuminuria in chronic kidney disease: a prospective controlled study. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association 2014; 29: 97-109. Pigeon C, Ilyin G, Courselaud B et al: A new mouse liver-specific gene,

118.

encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. The Journal of biological chemistry 2001; 276: 7811-9. Wrighting DM, Andrews NC: Interleukin-6 induces hepcidin expression

121.

122.

123.

124.

NU

MA

D

TE

CE P

120.

through STAT3. Blood 2006; 108: 3204-9. Verga Falzacappa MV, Vujic Spasic M, Kessler R et al: STAT3 mediates hepatic hepcidin expression and its inflammatory stimulation. Blood 2007; 109: 353-8. Babitt JL, Lin HY: The molecular pathogenesis of hereditary hemochromatosis. Seminars in liver disease 2011; 31: 280-92. Nemeth E, Tuttle MS, Powelson J et al: Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004; 306:

AC

119.

SC R

IP

115.

T

114.

2090-3. Babitt JL, Lin HY: Molecular mechanisms of hepcidin regulation: implications for the anemia of CKD. American journal of kidney diseases : the official journal of the National Kidney Foundation 2010; 55: 726-41. Gaweda AE, Goldsmith LJ, Brier ME, Aronoff GR: Iron, inflammation, dialysis adequacy, nutritional status, and hyperparathyroidism modify erythropoietic response. Clinical journal of the American Society of Nephrology : CJASN 2010; 5: 576-81. Kalantar-Zadeh K, Lee GH, Miller JE et al: Predictors of hyporesponsiveness to erythropoiesis-stimulating agents in hemodialysis patients. American journal of kidney diseases : the official journal of the National Kidney Foundation 2009; 53: 823-34.

ACCEPTED MANUSCRIPT

Association 2013; 28: 1672-9. Albitar S, Genin R, Fen-Chong M et al: High-dose alfacalcidol improves anaemia in patients on haemodialysis. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association European Renal Association 1997; 12: 514-8.

130.

Kiss Z, Ambrus C, Almasi C et al: Serum 25(OH)-cholecalciferol concentration is associated with hemoglobin level and erythropoietin resistance in patients on maintenance hemodialysis. Nephron Clinical practice 2011; 117: c373-8. Baeke F, Takiishi T, Korf H, Gysemans C, Mathieu C: Vitamin D: modulator of

D

129.

MA

NU

128.

T

127.

IP

126.

Patel NM, Gutierrez OM, Andress DL et al: Vitamin D deficiency and anemia in early chronic kidney disease. Kidney international 2010; 77: 715-20. Perlstein TS, Pande R, Berliner N, Vanasse GJ: Prevalence of 25-hydroxyvitamin D deficiency in subgroups of elderly persons with anemia: association with anemia of inflammation. Blood 2011; 117: 2800-6. Icardi A, Paoletti E, De Nicola L et al: Renal anaemia and EPO hyporesponsiveness associated with vitamin D deficiency: the potential role of inflammation. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal

SC R

125.

AC

CE P

TE

the immune system. Current opinion in pharmacology 2010; 10: 482-96. 131. Chow EC, Magomedova L, Quach HP et al: Vitamin D receptor activation down-regulates the small heterodimer partner and increases CYP7A1 to lower cholesterol. Gastroenterology 2014; 146: 1048-59. 132. Gupta S, Pandak WM, Hylemon PB: LXR alpha is the dominant regulator of CYP7A1 transcription. Biochemical and biophysical research communications 2002; 293: 338-43. 133. Chiang JY: Bile acids: regulation of synthesis. Journal of lipid research 2009; 50:

136.

1955-66. Jelinek DF, Andersson S, Slaughter CA, Russell DW: Cloning and regulation of cholesterol 7 alpha-hydroxylase, the rate-limiting enzyme in bile acid biosynthesis. The Journal of biological chemistry 1990; 265: 8190-7. Honjo Y, Sasaki S, Kobayashi Y, Misawa H, Nakamura H: 1,25-dihydroxyvitamin D3 and its receptor inhibit the chenodeoxycholic acid-dependent transactivation by farnesoid X receptor. The Journal of endocrinology 2006; 188: 635-43. Jiang W, Miyamoto T, Kakizawa T et al: Inhibition of LXRalpha signaling by

137.

vitamin D receptor: possible role of VDR in bile acid synthesis. Biochemical and biophysical research communications 2006; 351: 176-84. Singer RF: Vitamin D in dialysis: defining deficiency and rationale for

134.

135.

ACCEPTED MANUSCRIPT

141.

postmenopausal women: a 2013 update of the 2008 recommendations from the European Society for Clinical and Economic Aspects of Osteoporosis and Osteoarthritis (ESCEO). Current medical research and opinion 2013; 29: 305-13. Saliba W, Barnett O, Rennert HS, Lavi I, Rennert G: The relationship between

138.

NU

MA

serum 25(OH)D and parathyroid hormone levels. The American journal of medicine 2011; 124: 1165-70. Gomez-Alonso C, Naves-Diaz ML, Fernandez-Martin JL et al: Vitamin D status and secondary hyperparathyroidism: the importance of 25-hydroxyvitamin D

D

142.

SC R

IP

139.

T

140.

supplementation. Seminars in dialysis 2013; 26: 40-6. Zisman AL, Hristova M, Ho LT, Sprague SM: Impact of ergocalciferol treatment of vitamin D deficiency on serum parathyroid hormone concentrations in chronic kidney disease. American journal of nephrology 2007; 27: 36-43. Jean G, Terrat JC, Vanel T et al: Evidence for persistent vitamin D 1-alpha-hydroxylation in hemodialysis patients: evolution of serum 1,25-dihydroxycholecalciferol after 6 months of 25-hydroxycholecalciferol treatment. Nephron Clinical practice 2008; 110: c58-65. Rizzoli R, Boonen S, Brandi ML et al: Vitamin D supplementation in elderly or

145.

146.

147.

148.

TE

CE P

144.

AC

143.

cut-off levels. Kidney international Supplement 2003: S44-8. Ramirez JA, Emmett M, White MG et al: The absorption of dietary phosphorus and calcium in hemodialysis patients. Kidney international 1986; 30: 753-9. Dusso A, Gonzalez EA, Martin KJ: Vitamin D in chronic kidney disease. Best practice & research Clinical endocrinology & metabolism 2011; 25: 647-55. Kandula P, Dobre M, Schold JD et al: Vitamin D supplementation in chronic kidney disease: a systematic review and meta-analysis of observational studies and randomized controlled trials. Clinical journal of the American Society of Nephrology : CJASN 2011; 6: 50-62. Jean G, Vanel T, Terrat JC, Chazot C: Prevention of secondary hyperparathyroidism in hemodialysis patients: the key role of native vitamin D supplementation. Hemodialysis international International Symposium on Home Hemodialysis 2010; 14: 486-91. Jean G, Terrat JC, Vanel T et al: Daily oral 25-hydroxycholecalciferol supplementation for vitamin D deficiency in haemodialysis patients: effects on mineral metabolism and bone markers. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association 2008; 23: 3670-6. Vondracek SF, Hoody DW: Combination vitamin D therapy in stage 5 chronic

ACCEPTED MANUSCRIPT

T

IP

SC R

MA

NU

presenting as acute renal failure. Indian journal of nephrology 2008; 18: 125-6.

D

152.

TE

151.

CE P

150.

AC

149.

kidney disease. The Annals of pharmacotherapy 2011; 45: 1011-5. Aloia JF: Clinical Review: The 2011 report on dietary reference intake for vitamin D: where do we go from here? The Journal of clinical endocrinology and metabolism 2011; 96: 2987-96. McNally JD, Iliriani K, Pojsupap S et al: Rapid normalization of vitamin D levels: a meta-analysis. Pediatrics 2015; 135: e152-66. Ozkan B, Hatun S, Bereket A: Vitamin D intoxication. The Turkish journal of pediatrics 2012; 54: 93-8. Naik MA, Banday KA, Najar MS, Reshi AR, Bhat MA: Vitamin D intoxication

AC

CE P

TE

D

MA

NU

SC R

IP

T

ACCEPTED MANUSCRIPT

Figure 1

AC

CE P

TE

D

MA

NU

SC R

IP

T

ACCEPTED MANUSCRIPT

Figure 2

MA

NU

SC R

IP

T

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

Figure 3

AC

CE P

TE

D

MA

NU

SC R

IP

T

ACCEPTED MANUSCRIPT

Figure 4

ACCEPTED MANUSCRIPT

SC R

IP

T

Figure 1. Vitamin D metabolism and its regulation Dermal synthesis of vitamin D (cholecalciferol) from cholesterol depends on UV exposure (UV-B radiation), which is the major natural source of vitamin D. Because vitamin D from the diet or dermal synthesis is biologically inactive, it must be converted to its active form 1,25(OH)2D or calcitriol by the liver enzyme 25-hydroxylase and renal enzyme 1-α-hydroxylase. The mitochondrial protein 24-hydroxylase initiates the degradation of 25(OH)D and 1,25(OH)2D through hydroxylation of the side chain to form calcitroic acids. CKD, diabetes mellitus, increased FGF-23, and active vitamin D supplementation reduce the activity of

MA

NU

25-hydroxylase and 1-α-hydroxylase and increase the activity of 24-hydroxylase. This reduces endogenous 25(OH)D and 1,25(OH)2D production and enhances their degradation. CKD patients with secondary hyperparathyroidism on high doses of active vitamin-D may have aggravated 25(OH)D deficiency, which may also reduce the availability of 25(OH)D in extrarenal tissues, organs, and cells such as the skin,

TE

D

prostate, colon, brain, breast, lungs, placenta, osteoblasts, parathyroid gland, pancreas, muscles, monocytes, and T/B cells.

AC

CE P

Figure 2. Native vitamin D and bone turnover disorders In patients with secondary hyperparathyroidism (high bone turnover), native vitamin D supplements increase the production of 1,25(OH)2D in the parathyroid gland through autocrine or paracrine mechanisms that suppress the activity of the chief cells producing the parathyroid hormone. The combination therapy of native vitamin D and active vitamin D supplements has fewer adverse effects of hypercalcemia and hyperphosphatemia and can improve bone quality efficiently. In patients with adynamic bone disorder (low bone turnover), the viability of osteoblasts and osteoclasts is low. Providing native vitamin D or intermittent PTH supplements may rescue the function of osteoblasts, improve bone turnover, and promote bone health.

Figure 3. Influence of vitamin D on bone–fat connection Adipocytes and osteoblasts originate from a common mesenchymal precursor (MSCs) that can differentiate into other cell types; however, among the various cell fates, differentiation into adipocytes or osteoblasts is of particular relevance because the factors that enable osteoblastogenesis inhibit adipogenesis and vice versa. Noggin

ACCEPTED MANUSCRIPT

SC R

IP

T

induces adipogenic differentiation of MSCs through a novel mechanism. People with a high body mass index have elevated circulating noggin levels in plasma. In addition to inhibiting the osteoblast differentiation of MSCs, noggin may induce adipogenesis. Alternatively, activation of PPAR-γ promotes the differentiation of MSCs into adipocytes over osteoblasts. By contrast, the Wnt signaling pathway inhibits adipogenesis while supporting osteogenesis. These two pathways can also influence each other. Wnt signaling negatively regulates adipogenesis through β-catenin, which inhibits PPAR-γ-induced genes. Wnt10b, one of the Wnt family members, plays a key role in bone formation. The induction of Wnt10b-mediated osteogenesis

MA

NU

is caused by its ability to inhibit PPAR-γ expression. Adequate vitamin D supplements not only enhance osteoblast differentiation and function but also induce adipocyte apoptosis, resulting in increased bone mass and a reduced likelihood of osteoporosis development.

D

Figure 4. Vitamin D deficiency and anemia in CKD CKD patients often present with increased serum markers of inflammation, anemia

AC

CE P

TE

and EPO resistance. 25(OH)D and 1,25 (OH)2D deficiencies alter innate and adaptive immune function, which will increase inflammatory cytokine production (IL-6, IFN-γ, TFN-α) which will stimulate canonical janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, and phosphorylation of JAKs and STAT3. Phosphorylated-STAT3 homodimers enter the nucleus and bind to the hepcidin promoter to stimulate hepcidin expression in liver. The liver is the major source of hepcidin production. Inflammation stimulates, whereas erythropoiesis and growth factors decrease, hepcidin production. Hepcidin downregulates ferroportin in macrophages, enterocytes, and hepatocytes, leading to decreased iron release into the serum that is subsequently bound to transferrin for erythropoiesis. In addition, secondary hyperparathyroidism will directly inhibit erythroid progenitors, endogenous EPO synthesis, and RBC survival. It also indirectly promotes bone marrow fibrosis, hyperphosphataemia, and increase serum alkaline phosphatase. All of them will lead to ESA hyporesponsiveness.

ACCEPTED MANUSCRIPT Table 1. Human cells containing 1-α-hydroxylase and vitamin D receptor

Brain cells



Decidual stromal cells



Dendritic cells



Macrophages



T or B cells



Fetal trophoblasts



Osteoblasts



Osteoclasts



Keratinocytes



Breast cells



Prostate cells



Pancreas cells



Colon cells



Renal tubular cells



Enterocytes



Vascular endothelial cells

SC R NU MA

D

TE

CE P AC

IP



T

Human cells containing 1-α-hydroxylase and vitamin D receptor

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

Highlights  CKD patients are prone to deficiencies in both 25(OH)D and 1,25(OH)2D.  Circulating 25(OH)D can convert to 1,25(OH)2D in the extra-renal tissues.  Native vitamin D improves skeletal quality and quantity in CKD patients.  Native vitamin D promotes extra-skeletal health in CKD patients.  The benefit of combination of native and active vitamin D therapy in CKD.

Pleiotropic effects of vitamin D in chronic kidney disease.

Low 25(OH)D levels are common in chronic kidney disease (CKD) patients and are implicated in all-cause mortality and morbidity risks. Furthermore, the...
564B Sizes 1 Downloads 13 Views