Вы находитесь на странице: 1из 7

Nephrology 23, Suppl.

4 (2018) 88–94

Review Ar ticle

Mineral bone disorders in chronic kidney disease


YI-CHOU HOU,1 CHIEN-LIN LU2 and KUO-CHENG LU2
1
Department of Internal Medicine, Cardinal Tien Hospital, School of Medicine, Fu-Jen Catholic University, and 2Department of Medicine, Fu-Jen Catholic
University Hospital, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan

KEY WORDS: ABSTRACT:


renal osteodystrophy, vascular calcification,
vitamin D deficiency. As the GFR loss aggravates, the disturbed mineral metabolism worsens the
bone microstructure and remodelling – scenario, which is known as CKD-
Correspondence mineral bone disease (MBD). CKD-MBD is characterized by : (i) abnormal
Dr Kuo-Cheng Lu, Department of Medicine, Fu- metabolism of calcium, phosphorus, parathyroid hormone (PTH), or vitamin
Jen Catholic University Hospital, College of
D; (ii) abnormalities in bone turnover, mineralization, volume linear growth
Medicine, Fu-Jen Catholic University, New Taipei
or strength; (iii) soft-tissue calcifications, either vascular or extra-osseous.
City, Taiwan. Email: kuochenglu@gmail.com
Uremic vascular calcification and osteoporosis are the most common compli-
Accepted for publication 10 August 2018. cations related to CKD-MBD. Disregulated bone turnover by uremic toxin or
secondary hyperparathyroidism disturbed bone mineralization and makes it
doi: 10.1111/nep.13457 difficult for calcium and inorganic phosphate to enter into bone, resulting in
increased serum calcium and inorganic phosphate. Vascular calcification
worsens by hyperphosphatemia and systemic inflammation. Since vitamin D
deficiency plays an important role in renal osteodystrophy, supplement of
nutritional vitamin D is important in treating uremic osteoporosis and vas-
SUMMARY AT A GLANCE
cular calcification at the same time. Its pleotropic effect improves the bone
This article is a systematic review of the remodeling initiated by osteoblast and alleviates the risk factors for vascular
mechanism of CKDMBD and the calcification with less hypercalcemia than vitamin D receptor analogs. There-
complication about CKDMBD, including fore, nutritional vitamin D should be considered in managing CKDMBD.
uremic vascular calcification and
osteoporosis. Nutritional vitamin D, should
play a role in managing CKDMBD.

Physiological bone remodelling is an ever-lasting and deli- Kidney Disease: Improving Global Outcomes (KDIGO)
cately coordinated process between bone formation and defined CKD-MBD as a broader systemic disorder of mineral
resorption. It is comprised of continuous removal of old and bone metabolism as a result of CKD.4 CKD-MBD is
bone, replacing them with a newly synthesized proteina- characterized by the following: (i) abnormal metabolism of
ceous matrix, and to form new bone by subsequent mineral- calcium, phosphorus, parathyroid hormone (PTH), or vita-
ization of the matrix.1 Bone remodelling started with bone min D; (ii) abnormalities in bone turnover, mineralization,
resorption with osteoclast (OC) activation2 and then bone volume linear growth or strength; (iii) soft-tissue calcifica-
formation. Both resorption and formation are essential for tions, either vascular or extra-osseous.5
repairing micro-fractures and for modifying bone structure
as a stress response.3 In other words, remodelling is the
CHARACTERISTICS OF MINERAL AND
replacement of bone without changing its previous shape.
HORMONAL DISRUPTION IN CKD
Bone turnover occurs in both cortical and trabecular bone
with a relative higher turnover rate in trabecular bone. As the GFR falls, free serum calcium levels fall and serum
At the onset of chronic kidney disease (CKD), the sys- phosphorus increases. Because of GFR loss, compensatory
temic mineral metabolism and bone composition start to production of fibroblast growth factor 23 (FGF-23) decreases
change along with glomerular filtration rate (GFR) loss. As levels of sodium-dependent phosphate transport protein
the GFR loss aggravates, the disturbed mineral metabolism (Npt)2a and Npt2c in the kidney, resulting in increased uri-
worsens the bone microstructure and remodelling – scenario nary excretion of phosphate.6 In response, the parathyroid
that is known as CKD-mineral bone disease (MBD). The glands increase the production of PTH, which decreases the

88 © 2018 Asian Pacific Society of Nephrology


Mineral bone disorders in CKD

abundance of Npt2a and Npt2c in the proximal tubule, lead- proteinuria.25 As GFR loss, the production of FGF-23
ing to increased urinary Pi excretion that in turn lowers inhibits the 1-α-hydroxylase activity in the renal proximal
serum Pi levels.7 FGF23 also inhibits the production of 1,25 tubule.8 The damaged tubulointerstitial tissue reduced the
(OH)2D and therefore decreases intestinal Pi absorption, fur- activity of 1-α-hydroxylase and 25-hydroxylase.26 SPTH
ther decreasing serum Pi levels.8 The decreased 1,25(OH)2D itself also inhibits the hepatic production of 25(OH)D. Low
induces hypocalcaemia and then stimulated PTH production serum 25(OH)D concentration is common in CKD and
persists, which ensues secondary hyperparathyroidism ESRD patients,27–29 and the severity of 25(OH)D deficiency
(SHPT).9 As GFR continues to fall, however, these compensa- is related to the circulating PTH concentration.30,31 Michaud
tory mechanisms fail, leading to hyperphosphatemia, hyper- et al.32 reported that the SPTH reduced the hepatic produc-
parathyroidism, and higher serum FGF-23 concentration. The tion of calcidiol. Segersten et al.33 demonstrated 1α-OHase
SHPT and uremic toxin accumulation accelerate bone turn- expression in parathyroid glands, which suppressed parathy-
over by activating osteoclastogenesis and increased the roid gland hyperplasia in an autocrine/paracrine manner.
release of calcium and phosphate from bone.10 Under physio- Therefore, active vitamin D has been applied as the essential
logical condition, FGF-23 decreases the PTH from gland.11 As treatment in treating SHPT.34,35 However, evidences had
progressive GFR loss, PTH was resistant to the suppression by shown that supra-physiological or pharmacological dosage
FGF-23 and sequential nodular parathyroid hyperplasia of active 1,25(OH)2D may aggravate 25(OH)D3 deficiency
formed.12,13 On the aspect of vascular health, hyperphospha- because of its feedback inhibition of hepatic 1α-OHase and
temia, SHPT, and hypovitaminosis were strongly associated 25α-OHase resulting in 25(OH)D3 shortage in other tissues
with increased cardiovascular morbidity and mortality and or organs (e.g., colon, osteoblasts (OB), parathyroid gland,
increases the risk of calciphylaxis.14 Therefore, the treatment monocytes and immune cells, etc.).36 Active 1,25(OH)2D
on SHPT focus on (i) phosphate restriction, (ii) calcimimetics inactivates the 1α-OHase through the epigenetic modifica-
and (iii) vitamin D analogues supplement.4 tion of VDR gene.37 At the same time, the raising of intracel-
Hyperphosphatemia is associated with higher mortality in lular 1,25-(OH)2D3 activates the 24-OHase activity to
CKD/end-stage renal disease (ESRD) patients,15 and phos- autoregulates its own catabolism.38 Thus, active 1,25(OH)2D
phate restriction is mandatory in treating SHPT, and both may not only reduce 25(OH)D3 production but increase
dietary restriction and oral phosphate binders are important 1,25(OH)2D and 25(OH)D3 catabolism. Our study group
strategies. Among the phosphate binders, calcium-based provided the evidence that the haemodialysis (HD) patients
phosphate binders hindered the effect of vitamin D because had better control on SHPT with combination therapy with
of the risk of hypercalcemia.16 Calcium-free phosphate vitamin D analogue and cholecalciferol than those with vita-
binders such as sevelamer or lanthanum decreases the min D analogue alone, and the patients with combination
intestinal calcium loading, and it provided the benefit in therapy had higher cathelicidin concentration.39 Therefore,
mortality in contrast to the calcium-based phosphate supplement of 25(OH)D during the treatment of SHPT is
binder.17,18 The calcium-sensing receptor (CaSR) is impor- important when prescribing active vitamin D or vitamin D
tant in regulating PTH secretion and therefore, this target analogue.
offers the potential to suppress PTH secretion by comple-
mentary mechanism to vitamin D analogues. Calcimimetics
VASCULAR AND SOFT TISSUE
allosterically enhance the sensitivity of CaSR in the parathy-
CALCIFICATION IN CKD
roid glands to calcium.19 As it direct suppression on PTH,
the serum calcium and phosphate concentration could be In CKD, VC will be present at the beginning stage of CKD
controlled. Current studies involving the use of cinacalcet because of the impaired bone mineral metabolism.40 VC can
include ADVANCE and Evaluation of Cinacalcet HCl Ther- be considered as two major types: intimal calcification asso-
apy to Lower Cardiovascular Events (EVOLVE) studies, and ciated with atherosclerosis, and medial calcification that
the use of calcimimetics provided the benefits on controlling involves damaged vascular smooth muscle cells (VSMC),
vascular calcification (VC) in subgroup analysis.20–22 leading to increased vascular stiffness and decreased vascu-
lar elasticity.41 The factors involving the pathophysiology of
VC in CKD include abnormal serum calcium and phosphate
NOVEL CONCEPTS OF ROLE OF
levels, hyperparathyroidism, increased matrix degradation,
NUTRITIONAL VITAMIN D IN SHPT
VSMC apoptosis, decreased glutamate protein in matrix, and
Traditionally, active vitamin D compounds were used in systemic inflammation.42–45 The factors above-mentioned
SHPT patients for its inhibitory effects on PTH gene tran- activated the osteoblastic transformation of VSMC and
scription and chief cell hyperplasia.23 As CKD progresses, the VC.
vitamin D deficiency or insufficiency was prominent because These will make way to let VSMC transdifferentiate to
of the presence of proteinuria, tubulointerstitial injury and phenotypic osteoblastic cells. In addition, patients with CKD
GFR loss.24 The binding of 25(OH)D to the proximal tubular usually have bone turnover problems.46 In a high turnover
epithelium megalin receptor is limited during the status, SHPT increases calcium and phosphate release from

© 2018 Asian Pacific Society of Nephrology 89


Y-C Hou et al.

bone by activating bone resorption.47 However, in a low turnover status in SHPT can induce increased bone deminer-
turnover status (such as adynamic bone disorder), circulat- alization, which will increase calcium and inorganic phos-
ing phosphate and calcium cannot efficiently deposit into phate release from bone into circulation. In contrast,
the bone surface and this might maintain serum calcium overtreatment of CKD patients with Ca-salts, vitamin D
and phosphate levels to a higher levels.48 Consequently, cir- receptor analogue (VDRA), or aluminium may cause them
culating phosphate and calcium tend to deposit in soft tissue to develop low turnover bone disorders and low serum PTH
other than bone matrix, causing higher probability of levels. The decreased bone mineralization makes it difficult
VC. Interestingly, during the VC process, the calcifying for calcium and inorganic phosphate to enter into bone,
VSMC will secrete sclerostin (SOST), a Wingless/Int-1 resulting in increased serum calcium and inorganic phos-
(Wnt)-signalling inhibitor that may not only counteract the phate.58 Both high and low bone turnover disorders are
local calcification in the artery wall but also destroy bone characterized by a relatively higher degree of bone resorp-
mineralization.49,50 These phenomena may lead to reduced tion than bone formation, which may contribute to the ele-
bone mass with a vicious cycle between bone turnover vated serum calcium and inorganic phosphate levels, and
and VC. aggravate VC/ossification.

TRADITIONAL FACTORS RELATED TO THE NOVEL CONCEPTS OF NUTRITIONAL


OSTEOGENIC TRANS-DIFFERENTIATION OF VITAMIN D IN VASCULAR CALCIFICATION
VASCULAR SMOOTH MUSCLE CELLS IN CKD IN CKD
As GFR decline, the secretion of FGF-23 from osteocytes In the study of cardiovascular disease with childhood onset
increases to augment the renal excretion of phosphate.8 of ESRD, the Berlin paediatricians Briese et al.59 pointed out
However, FGF-23 inhibits the activity of angiotensin- interesting differences (with a relatively similar study result
converting enzyme 2 (ACE2). FGF-23 blocks the conversion reported by Pilz et al.60): the prevalence of coronary calcifi-
of angiotensin-II into angiotensin (1–7).51 The increase in cations (10% vs 92%) and of cardiac valve calcifications
FGF23 disturbs the renin-angiotensin aldosterone system (0% vs 32%) was quite lower in the Berlin study (more use
(RASS) system and lessens the angiotensin receptor blocker of cholecalciferol) than in the Heidelberg one (less use of
(ARB) efficacy.52 The activated angiotensin-II will enhance cholecalciferol). In contrast to active vitamin D, the inci-
the production of aldosterone. Phosphate and calcium stim- dence of hypercalcaemia is less in CKD patients,61,62 and it
ulate the Na-Pi cotransporter, and aldosterone also contrib- might avoid extra-osseous calcium deposition in the vessels.
utes to activate the Na-Pi cotransporter, resulting in These findings suggest that nutritional vitamin D supple-
increased phosphate entrance into VSMC.53,54 In addition, ment may prevent the development of VC.
aldosterone accentuates the inflammatory status in part by
tumour necrosis factor alpha (TNFα). Both oxidative/inflam-
VITAMIN D DEFICIENCY MAY CONTRIBUTE
matory status and increased intracellular phosphate levels
TO VASCULAR CALCIFICATION THROUGH
promote VSMC to transdifferentiate into phenotypic OB
INCREASE IN GLI1 EXPRESSION IN CKD
cells, which causes the ossification of the vascular wall to
progress.54 As a whole, the calcified vessels have more Mesenchymal stem cell (MSC)-like cells reside in the vascu-
prominent bone formation characteristic than bone resorp- lar wall, especially in the inner layer of tunica adventitia.
tion ones. In addition, osteogenic cells may secrete SOST Perivascular Gli1+ progenitors are key contributors to
and FGF23. The secreted FGF23 from the calcified vessel injury-induced organ fibrosis.63 Kramann et al.64 provided
may contribute further increased FGF23 serum levels. These the evidence that Gli1+ adventitial cells have a critical role
high serum levels of FGF23, SOST will contribute to in VC in CKD. It has been noticed that both active and
decreasing OB function with resulted in further decrease nutritional vitamin D serve as the suppressor of Gli1 gene.
bone turnover in CKD patients. Vitamin D activated receptor regulates specific microRNA
and secrets hedgehog (Hh) pathway inhibitors suppressor of
fused (SuFu).65 Cholecalciferol could repress the Hh signal-
LINKAGE BETWEEN BONE TURNOVER AND
ling by directly inhibiting extra-cellular domain of smooth-
VC IN CKD
ened.66,67 The vitamin D deficiency may contribute to VC
Basically, bone cells have less vitality in patients with CKD through increase in Gli1 expression in CKD.
than in normal persons because of accumulation of uremic
toxin and insulin resistance. Thus, low bone turnover is part
BONE DISORDERS IN CKD
of the innate character of CKD.55–57 High PTH serum levels
will overcome the indolent bone cells and lead to high turn- In patients with early CKD (stages 1 and 2), Wnt-signalling
over bone disease with the characteristics of relatively inhibitors, such as dickkopf WNT-signalling pathway inhibi-
higher bone resorption than bone resorption.7 The high tor 1 (DKK1), SOST and secreted frizzled related protein 1

90 © 2018 Asian Pacific Society of Nephrology


Mineral bone disorders in CKD

(sFRP) were secreted from kidneys, or osteocyte in bone/cal- decrease gastrointestinal absorption and increase clearance
cified soft tissue, which could act on the OBs resulting in the uremic toxin may be important in treating the uremic
decreased OB viability.68,69 In addition, retention of protein- osteoporosis.
bound uremic toxins (PBUT), such as indoxyl sulphate/p-
cresol sulphate (IS/PCS), further attenuated the OB/OC via-
RENAL OSTEODYSTROPHY
bility and function.55,70 Uremic osteoporosis was used to
describe the effects of PBUT on the qualitative bone loss The prevalence of osteoporosis varies according to CKD
with normal bone quantity. When renal function declined stages. Fractures in early-stage CKD patients (stages 1–3 A
progressively, metabolic acidosis and hyponatremia also CKD) are more resemblance as traditional osteoporosis
result in the bone quantity loss.71 When the severity of CKD rather than CKD-MBD.85,86 However, most patients in stage
progresses (≥stage 3) with calcium, phosphate, vitamin D 4 or 5 CKD exhibit certain degree of decreased bone mineral
and PTH dysregulation, patients may present with high or density (BMD) and/or certain level of CKD-MBD.87,88 At
low PTH levels. High PTH levels drive the indolent OB into the time of dialysis initiation, as many as up to half of the
high viability and function but poor quality in behaviour, patients, might have experienced a fracture.89 Together,
resulting in both bone quality and quantity loss.72,73 How- vitamin D deficiency, poor nutrition, inactivity, myopathy,
ever, medical or surgical treatment of SHPT which largely and peripheral neuropathy play a role in muscle weakness
remove much of the PTH hypersecretion, the bone cells may and falls.90 Since vitamin D is crucial for bone health, and
return back to the innate low bone cell viability status –the vitamin D supplement is protective in fracture prevention in
low bone turnover disorders.74,75 CKD, the role of vitamin D should be stratified.

UREMIC OSTEOPOROSIS-RELATED BONE VITAMIN D DEFICIENCY-RELATED BONE


QUALITY LOSS QUANTITY LOSS
End-stage renal disease patients also are at an extremely Vitamin D is essential for normal bone formation and miner-
high risk of bone fractures because of the high incidence of alization, and plays a critical role in bone biology.91,92 Disor-
uremic osteoporosis and MBD.76,77 Compared with normal dered vitamin D metabolism plays a crucial role in SHPT
population, uremic patients are more prone to having along with other changes of mineral metabolism. Vitamin D
abnormal bone metabolism, a disarranged bone microarchi- deficiency is common in advanced CKD, and the BMD is pos-
tecture, lower bone mass and musculoskeletal fragility.78,79 itively correlated with the serum 25(OH)D concentration.93
The concept of ‘uremic osteoporosis’ was recently intro- In another study, Mucsi et al.94,95 demonstrated a correlation
duced by Fukagawa et al.5,80 to explain the role of uremic between 25 (OH) vitamin D deficiency and low radial BMD
toxins in influencing bone quality in CKD patients. Accumu- in HD patients. ESRD patients with 25 (OH) D deficiency
lative uremic toxins exhibit noxious effects on bone metabo- (<15 nmol/L) had a lower bone formation rate and decreased
lism and function, and attenuate the bone quality and trabecular mineralization surface, independent of PTH and
quantity. A representative uremic toxin, IS, accumulates in calcitriol levels.95,96 Vitamin D can suppress the parathyroid
CKD patients with progressive renal dysfunction.81 IS could gland, and vitamin D deficiency worsens SHPT and increased
down-regulate the expression of PTH receptor on OB and bone turnover, bone loss, and mineralization defects. Mili-
decrease the bone turnover.82 It also represses the bone for- nkovic et al.97 have been noted the bone resorption increased
mation signalling by enhancing the secretion of Wnt antago- in HD patients with serum 25-(OH) D levels <50 nmol/L In a
nist from osteocyte.57 A previous clinical study showed a retrospective study, Ambrus et al.95 revealed the association
positive correlation between IS levels and bone formation between fractures and significant low vitamin D status among
rate, osteoid volume, OB surface area and bone fibrosis vol- maintenance HD patients. Lower 25 (OH) vitamin D levels
ume. Multivariate regression analysis further confirmed the have been shown to be associated with subperiosteal resorp-
associations between IS levels and bone formation rate, OB tion and reduced BMD in ESRD patients. Vitamin D defi-
surface area, and bone fibrosis volume.83 They revealed that ciency might also cause frailty presenting muscle weakness,
the mineral-to-matrix ratio and carbonate substitution non-vertebral and hip fractures.90,98,99 Although high-quality
increased with the number of immature crystals in CKD studies are needed to validate the effect of vitamin D on bone
rats.10 The enzymatic cross-link ratio was also increased quantity in CKD, supplement of vitamin D is important in
abnormally. Accumulated uremic toxins, especially IS, might treating quantity bone loss.
also be responsible for pathological collagen cross-links with
disarrayed orientation and weakened mechanical proper-
VITAMIN D DEFICIENCY-RELATED BONE
ties.84 Although the benefit of oral absorbent AST-120 for
QUALITY LOSS
uremic osteoporosis is still lacking in the clinical studies, the
AST-120 significantly reduces serum IS levels and can abol- Vitamin D deficiency is associated with an impaired bone
ish the bone effects noted in CKD animals.10,84 Therefore, to mineralization process, resulting in a larger volume of the

© 2018 Asian Pacific Society of Nephrology 91


Y-C Hou et al.

unmineralized bone matrix, called osteoid. A group of US 8. Prie D, Friedlander G. Reciprocal control of 1,25-dihydroxyvitamin
and German scientists100,101 demonstrated that vitamin D D and FGF23 formation involving the FGF23/Klotho system. Clin.
J. Am. Soc. Nephrol. 2010; 5: 1717–22.
deficiency speeds up the premature aging of existing bone
9. Friedl C, Zitt E. Vitamin D prohormone in the treatment of
and consequently reduces its quality. In vitamin D defi- secondary hyperparathyroidism in patients with chronic kidney
ciency patients, the mineralized bone was composed of disease. Int. J. Nephrol. Renovasc. Dis. 2017; 10: 109–22.
higher degree of mature collagen cross-link ratio and 10. Iwasaki Y, Kazama JJ, Yamato H, Matsugaki A, Nakano T,
carbonate-to-phosphate ratio, characteristics of aging and Fukagawa M. Altered material properties are responsible for bone
fragility in rats with chronic kidney injury. Bone 2015; 81:
brittle bone. The premature mineralized bone has significant
247–54.
loss in the mechanical integrity and toughness, and the 11. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V et al. The parathyroid is
vitamin-D-deficient bone is less resistant to the crack initia- a target organ for FGF23 in rats. J. Clin. Invest. 2007; 117: 4003–8.
tion. Vitamin D-deficient bone tissue has a straighter crack 12. Tominaga Y, Takagi H. Molecular genetics of hyperparathyroid
path with fewer crack bridges compared with normal.101 disease. Curr. Opin. Nephrol. Hypertens. 1996; 5: 336–41.
13. Cunningham J, Locatelli F, Rodriguez M. Secondary
Furthermore, vitamin D deficiency inhibits normal bone
hyperparathyroidism: Pathogenesis, disease progression, and
remodelling in the remaining mineralized bone tissue.102,103 therapeutic options. Clin. J. Am. Soc. Nephrol. 2011; 6: 913–21.
Therefore, vitamin D levels should be maintained at well- 14. Finch JL, Lee DH, Liapis H et al. Phosphate restriction significantly
balanced levels to maintain the structural integrity of bone, reduces mortality in uremic rats with established vascular
especially in CKD patients. calcification. Kidney Int. 2013; 84: 1145–53.
15. Bellinghieri G, Santoro D, Savica V. Emerging drugs for
In conclusion, the underlying mechanisms of bone loss
hyperphosphatemia. Expert Opin. Emerg. Drugs 2007; 12: 355–65.
and fractures in CKD patients are complex and remain 16. Meric F, Yap P, Bia MJ. Etiology of hypercalcemia in hemodialysis
incompletely understood. In contrast to bone biopsy, the patients on calcium carbonate therapy. Am. J. Kidney Dis. 1990; 16:
presently available non-invasive investigative measures to 459–64.
detect both bone quantity and quality losses are clinically 17. Habbous S, Przech S, Acedillo R, Sarma S, Garg AX, Martin J. The
efficacy and safety of sevelamer and lanthanum versus calcium-
insufficient as screening or diagnostic techniques. Bone-
containing and iron-based binders in treating hyperphosphatemia
targeted pharmacotherapy was shown to only marginally in patients with chronic kidney disease: A systematic review and
affect the incidence of fractures, and might be associated meta-analysis. Nephrol. Dial. Transplant. 2017; 32: 111–25.
with unexpected adverse effects in this specific population. 18. Sekercioglu N, Thabane L, Diaz Martinez JP et al. Comparative
Classical but individualized therapy, including vitamin D effectiveness of phosphate binders in patients with chronic kidney
disease: A systematic review and network meta-analysis. PLoS One
supplementation, phosphate control with phosphate binders,
2016; 11: e0156891.
anti-resorptive agents, dialysis and medical and surgical PTX, 19. Valle C, Rodriguez M, Santamaria R et al. Cinacalcet reduces the set
might differentially benefit these patients. Researchers point of the PTH-calcium curve. J. Am. Soc. Nephrol. 2008; 19: 2430–6.
should keep on investigating the detailed mechanisms of 20. Raggi P, Chertow GM, Torres PU et al. The ADVANCE study: A
pathophysiology, and look for the targeted therapy for both randomized study to evaluate the effects of cinacalcet plus low-
dose vitamin D on vascular calcification in patients on
quality- and quantity-related bone losses in CKD patients.
hemodialysis. Nephrol. Dial. Transplant. 2011; 26: 1327–39.
21. The EVOLVE Trial Investigators. Effect of cinacalcet on
cardiovascular disease in patients undergoing dialysis. N. Engl.
J. Med. 2012; 367: 2482–94.
REFERENCES 22. Friedl C, Reibnegger G, Kramar R et al. Mortality in dialysis
patients with cinacalcet use: A large observational registry study.
1. Honma M, Ikebuchi Y, Kariya Y, Suzuki H. Regulatory Eur. J. Intern. Med. 2017; 42: 89–95.
mechanisms of RANKL presentation to osteoclast precursors. Curr. 23. Lips P. Vitamin D deficiency and secondary hyperparathyroidism
Osteoporos. Rep. 2014; 12: 115–20. in the elderly: Consequences for bone loss and fractures and
2. Parfitt AM. What is the normal rate of bone remodeling? Bone therapeutic implications. Endocr. Rev. 2001; 22: 477–501.
2004; 35: 1–3. 24. Hou Y-C, Liu W-C, Zheng C-M, Zheng J-Q, Yen T-H, Lu K-C. Role
3. Sai AJ, Walters RW, Fang X, Gallagher JC. Relationship between of vitamin D in uremic vascular calcification. Biomed. Res. Int. 2017;
vitamin D, parathyroid hormone, and bone health. J. Clin. 2017: 2803579.
Endocrinol. Metab. 2011; 96: E436–E46. 25. Christakos S, Ajibade DV, Dhawan P, Fechner AJ, Mady LJ. Vitamin
4. Lentine KL, Kasiske BL, Levey AS et al. KDIGO clinical practice D: Metabolism. Endocrinol. Metab. Clin. North Am. 2010; 39: 243–53.
guideline on the evaluation and care of living kidney donors. 26. Dusso AS, Brown AJ. Mechanism of vitamin D action and its
Transplantation 2017; 101: S7–S105. regulation. Am. J. Kidney Dis. 1998; 32: S13–24.
5. Fukagawa M, Komaba H. Chronic kidney disease-mineral and 27. Bhan I, Burnett-Bowie SAM, Ye J, Tonelli M, Thadhani R. Clinical
bone disorder in Asia. Kidney Dis. 2017; 3: 1–7. measures identify vitamin D deficiency in dialysis. Clin. J. Am. Soc.
6. Canalejo A, Almaden Y, De Smet R et al. Effects of uremic Nephrol. 2010; 5: 460–7.
ultrafiltrate on the regulation of the parathyroid cell cycle by 28. Nigwekar SU, Bhan I, Thadhani R. Ergocalciferol and
calcitriol. Kidney Int. 2003; 63: 732–7. cholecalciferol in CKD. Am. J. Kidney Dis. 2012; 60: 139–56.
7. Blaine J, Chonchol M, Levi M. Renal control of calcium, 29. Jean G, Souberbielle JC, Chazot C. Vitamin D in chronic kidney
phosphate, and magnesium homeostasis. Clin. J. Am. Soc. Nephrol. disease and dialysis patients. Nutrients 2017 Mar 25;9(4). pii: E328.
2015; 10: 1257–72. https://doi.org/10.3390/nu9040328.

92 © 2018 Asian Pacific Society of Nephrology


Mineral bone disorders in CKD

30. Kovesdy CP, Ahmadzadeh S, Anderson JE, Kalantar-Zadeh K. 50. Evenepoel P, D’Haese P, Brandenburg V. Sclerostin and DKK1:
Association of activated vitamin D treatment and mortality in New players in renal bone and vascular disease. Kidney Int. 2015;
chronic kidney disease. Arch. Intern. Med. 2008; 168: 397–403. 88: 235–40.
31. Levin A, Bakris GL, Molitch M et al. Prevalence of abnormal serum 51. Zanchi C, Locatelli M, Benigni A et al. Renal expression of FGF23
vitamin D, PTH, calcium, and phosphorus in patients with chronic in progressive renal disease of diabetes and the effect of ACE
kidney disease: Results of the study to evaluate early kidney inhibitor. PLoS One 2013; 8: e70775.
disease. Kidney Int. 2007; 71: 31–8. 52. de Jong MA, Mirkovic K, Mencke R et al. Fibroblast growth factor
32. Michaud J, Naud J, Ouimet D et al. Reduced hepatic synthesis of 23 modifies the pharmacological effects of angiotensin receptor
calcidiol in uremia. J. Am. Soc. Nephrol. 2010; 21: 1488–97. blockade in experimental renal fibrosis. Nephrol. Dial. Transplant.
33. Segersten U, Correa P, Hewison M et al. 25-hydroxyvitamin D(3)- 2017; 32: 73–80.
1alpha-hydroxylase expression in normal and pathological 53. Segawa H, Yamanaka S, Ito M et al. Internalization of renal type IIc
parathyroid glands. J. Clin. Endocrinol. Metab. 2002; 87: 2967–72. Na-pi cotransporter in response to a high-phosphate diet. Am.
34. Sprague SM, Llach F, Amdahl M, Taccetta C, Batlle D. Paricalcitol J. Physiol. Renal Physiol. 2005; 288: F587–F96.
versus calcitriol in the treatment of secondary 54. Lang F, Ritz E, Alesutan I, Voelkl J. Impact of aldosterone on
hyperparathyroidism. Kidney Int. 2003; 63: 1483–90. osteoinductive signaling and vascular calcification. Nephron Physiol.
35. Sprague SM, Silva AL, Al-Saghir F et al. Modified-release 2014; 128: 40–5.
calcifediol effectively controls secondary hyperparathyroidism 55. Watanabe K, Tominari T, Hirata M et al. Indoxyl sulfate, a uremic
associated with vitamin D insufficiency in chronic kidney disease. toxin in chronic kidney disease, suppresses both bone formation
Am. J. Nephrol. 2014; 40: 535–45. and bone resorption. FEBS Open Bio 2017; 7: 1178–85.
36. Bell NH, Shaw S, Turner RT. Evidence that 1,25-dihydroxyvitamin 56. Hamann C, Goettsch C, Mettelsiefen J et al. Delayed bone
D3 inhibits the hepatic production of 25-hydroxyvitamin D in regeneration and low bone mass in a rat model of insulin-resistant
man. J. Clin. Invest. 1984; 74: 1540–4. type 2 diabetes mellitus is due to impaired osteoblast function. Am.
37. Kouzmenko A, Ohtake F, Fujiki R, Kato S. Epigenetic J. Physiol. Endocrinol. Metab. 2011; 301: E1220–8.
modifications in vitamin D receptor-mediated transrepression. In: 57. Drueke TB, Massy ZA. Changing bone patterns with progression of
Feldman D, Pike JW, Adams, JS (eds). Vitamin D, 3rd edn. San chronic kidney disease. Kidney Int. 2016; 89: 289–302.
Diego,CA: Academic Press, 2011. 227–234. 58. Andress DL. Adynamic bone in patients with chronic kidney
38. Brenza HL, Kimmel-Jehan C, Jehan F et al. Parathyroid hormone disease. Kidney Int. 2008; 73: 1345–54.
activation of the 25-hydroxyvitamin D3-1alpha-hydroxylase gene 59. Briese S, Wiesner S, Will JC et al. Arterial and cardiac disease in
promoter. Proc. Natl. Acad. Sci. U. S. A. 1998; 95: 1387–91. young adults with childhood-onset end-stage renal disease-impact
39. Zheng JQ, Hou YC, Zheng CM et al. Cholecalciferol additively reduces of calcium and vitamin D therapy. Nephrol. Dial. Transplant. 2006;
serum parathyroid hormone and increases vitamin D and cathelicidin 21: 1906–14.
levels in Paricalcitol-Treated secondary hyperparathyroid 60. Pilz S, Verheyen N, Grubler MR, Tomaschitz A, Marz W. Vitamin
hemodialysis patients. Nutrients. 2016 Nov 5; 8(11). pii: E708. D and cardiovascular disease prevention. Nat. Rev. Cardiol. 2016;
40. Mafham M, Emberson J, Landray MJ, Wen C-P, Baigent C. Estimated 13: 404–17.
glomerular filtration rate and the risk of major vascular events and 61. Parikh C, Gutgarts V, Eisenberg E, Melamed ML. Vitamin D and
all-cause mortality: A meta-analysis. PLoS One 2011; 6: e25920. clinical outcomes in dialysis. Semin. Dial. 2015; 28: 604–9.
41. Shanahan CM, Crouthamel MH, Kapustin A, Giachelli CM. 62. Mazzaferro S, Goldsmith D, Larsson TE, Massy ZA, Cozzolino M.
Arterial calcification in chronic kidney disease: Key roles for Vitamin D metabolites and/or analogs: Which D for which patient?
calcium and phosphate. Circ. Res. 2011; 109: 697–711. Curr. Vasc. Pharmacol. 2014; 12: 339–49.
42. Wu M, Zhang JD, Tang RN et al. Elevated PTH induces endothelial- 63. Kramann R, Schneider RK, DiRocco DP et al. Perivascular Gli1+
to-chondrogenic transition in aortic endothelial cells. Am. J. Physiol. progenitors are key contributors to injury-induced organ fibrosis.
Renal Physiol. 2017; 312: F436–44. https://doi.org/10.1152/ Cell Stem Cell 2015; 16: 51–66.
ajprenal.00210.2016. 64. Kramann R, Goettsch C, Wongboonsin J et al. Adventitial MSC-
43. Hung SC, Kuo KL, Huang HL et al. Indoxyl sulfate suppresses like cells are progenitors of vascular smooth muscle cells and drive
endothelial progenitor cell-mediated neovascularization. Kidney Int. vascular calcification in chronic kidney disease. Cell Stem Cell 2016;
2016; 89: 574–85. 19: 628–42.
44. Abbasian N, Burton JO, Herbert KE et al. Hyperphosphatemia, 65. Alimirah F, Peng X, Gupta A et al. Crosstalk between the vitamin D
phosphoprotein phosphatases, and microparticle release in receptor (VDR) and miR-214 in regulating SuFu, a hedgehog pathway
vascular endothelial cells. J. Am. Soc. Nephrol. 2015; 26: 2152–62. inhibitor in breast cancer cells. Exp. Cell Res. 2016; 349: 15–22.
45. Evrard S, Delanaye P, Kamel S, Cristol JP, Cavalier E et al. Vascular 66. Cortes M, Liu SY, Kwan W, Alexa K, Goessling W, North TE.
calcification: From pathophysiology to biomarkers. Clin. Chim. Acta Accumulation of the vitamin D precursor cholecalciferol
2015; 438: 401–14. antagonizes hedgehog signaling to impair hemogenic endothelium
46. Lu KC, Wu CC, Yen JF, Liu WC. Vascular calcification and renal formation. Stem Cell Rep. 2015; 5: 471–9.
bone disorders. ScientificWorldJournal 2014; 2014: 637065. 67. Bijlsma MF, Spek CA, Zivkovic D, van de Water S, Rezaee F,
47. Almirall J, Gallardo X, Castane E. Effects of cinacalcet on vascular Peppelenbosch MP. Repression of smoothened by patched-
calcification in haemodialysis patients. Nephrol. Dial. Transplant. dependent (pro-)vitamin D3 secretion. PLoS Biol. 2006; 4: e232.
2010; 25: 2800. 68. Fang Y, Ginsberg C, Seifert M et al. CKD-induced wingless/
48. Davies MR, Lund RJ, Mathew S, Hruska KA. Low turnover integration1 inhibitors and phosphorus cause the CKD-mineral
osteodystrophy and vascular calcification are amenable to skeletal and bone disorder. J. Am. Soc. Nephrol. 2014; 25: 1760–73.
anabolism in an animal model of chronic kidney disease and the 69. Behets GJ, Viaene L, Meijers B et al. Circulating levels of sclerostin
metabolic syndrome. J. Am. Soc. Nephrol. 2005; 16: 917–28. but not DKK1 associate with laboratory parameters of CKD-MBD.
49. Zhu D, Mackenzie NCW, Millan JL, Farquharson C, MacRae VE. PLoS One 2017; 12: e0176411.
The appearance and modulation of osteocyte marker expression 70. Tanaka H, Iwasaki Y, Yamato H et al. P-Cresyl sulfate induces
during calcification of vascular smooth muscle cells. PLoS One osteoblast dysfunction through activating JNK and p38 MAPK
2011; 6: e19595. pathways. Bone 2013; 56: 347–54.

© 2018 Asian Pacific Society of Nephrology 93


Y-C Hou et al.

71. Bonjour JP. Nutritional disturbance in acid-base balance and not predict fragility fractures in men with moderate chronic kidney
osteoporosis: A hypothesis that disregards the essential disease. J. Clin. Densitom. 2016; 19: 146–53.
homeostatic role of the kidney. Br. J. Nutr. 2013; 110: 1168–77. 88. Nickolas TL, McMahon DJ, Shane E. Relationship between
72. Ketteler M, Block GA, Evenepoel P et al. Executive summary of the moderate to severe kidney disease and hip fracture in the United
2017 KDIGO chronic kidney disease-mineral and bone disorder States. J. Am. Soc. Nephrol. 2006; 17: 3223–32.
(CKD-MBD) guideline update: What’s changed and why it 89. Cosman F, Crittenden DB, Adachi JD et al. Romosozumab
matters. Kidney Int. 2017; 92: 26–36. treatment in postmenopausal women with osteoporosis. N. Engl.
73. Bover J, Bailone L, Lopez-Baez V et al. Osteoporosis, bone mineral J. Med. 2016; 375: 1532–43.
density and CKD-MBD: Treatment considerations. J. Nephrol. 2017; 90. Boudville N, Inderjeeth C, Elder GJ, Glendenning P. Association
30: 677–87. between 25-hydroxyvitamin D, somatic muscle weakness and falls
74. Hernandes FR, Canziani ME, Barreto FC et al. The shift from high to risk in end-stage renal failure. Clin. Endocrinol. (Oxf ) 2010; 73:
low turnover bone disease after parathyroidectomy is associated with 299–304.
the progression of vascular calcification in hemodialysis patients: A 91. Posa F, Di Benedetto A, Colaianni G et al. Vitamin D effects on
12-month follow-up study. PLoS One 2017; 12: e0174811. osteoblastic differentiation of mesenchymal stem cells from dental
75. Behets GJ, Spasovski G, Sterling LR et al. Bone histomorphometry tissues. Stem Cells Int. 2016; 2016: 9150819.
before and after long-term treatment with cinacalcet in dialysis 92. Takahashi N, Udagawa N, Suda T. Vitamin D endocrine system and
patients with secondary hyperparathyroidism. Kidney Int. 2015; 87: osteoclasts. Bonekey Rep. 2014; 3: 495.
846–56. 93. Elder GJ, Mackun K. 25-Hydroxyvitamin D deficiency and diabetes
76. Salam SN, Eastell R, Khwaja A. Fragility fractures and osteoporosis predict reduced BMD in patients with chronic kidney disease.
in CKD: Pathophysiology and diagnostic methods. Am. J. Kidney J. Bone Miner. Res. 2006; 21: 1778–84.
Dis. 2014; 63: 1049–59. 94. Ambrus C, Marton A, Nemeth ZK, Mucsi I. Bone mineral density
77. Wakasugi M, Kazama JJ, Wada A, Hamano T, Masakane I, in patients on maintenance dialysis. Int. Urol. Nephrol. 2010; 42:
Narita I. Hip fracture trends in Japanese dialysis patients, 723–39.
2008-2013. Am. J. Kidney Dis. 2018; 71: 173–81. 95. Ambrus C, Almasi C, Berta K et al. Vitamin D insufficiency and
78. Paranhos-Neto FP, Lima GAC, Silva LC et al. HR-pQCT detects bone fractures in patients on maintenance hemodialysis. Int. Urol.
alterations in bone microstructure in men with CKD stages 3 and Nephrol. 2011; 43: 475–82.
4, which are influenced by hormonal changes and body 96. Coen G, Mantella D, Manni M et al. 25-hydroxyvitamin D levels
composition. Clin. Nephrol. 2018; 2018: 10–7. and bone histomorphometry in hemodialysis renal osteodystrophy.
79. Malmgren L, McGuigan F, Christensson A, Akesson KE. Reduced Kidney Int. 2005; 68: 1840–8.
kidney function is associated with BMD, bone loss and markers of 97. Milinkovic N, Majkic-Singh NT, Mirkovic DD, Beletic AD,
mineral homeostasis in older women: A 10-year longitudinal Pejanovic SD, Vujanic ST. Relation between 25(OH)-vitamin D
study. Osteoporos. Int. 2017; 28: 3463–73. deficiency and markers of bone formation and resorption in
80. Yamamoto S, Fukagawa M. Uremic toxicity and bone in CKD. haemodialysis patients. Clin. Lab. 2009; 55: 333–9.
J. Nephrol. 2017; 30: 623–7. 98. Lee YH, Kim JE, Roh YH et al. The combination of vitamin D
81. Barreto FC, Barreto DV, Stinghen AEM, Massy ZA. Comment on deficiency and mild to moderate chronic kidney disease is
Indoxyl sulfate-review of toxicity and therapeutic strategies. Toxins associated with low bone mineral density and deteriorated femoral
2016, 8, 358. Toxins (Basel) 2017; 9: E142. microarchitecture: Results from the KNHANES 2008-2011. J. Clin.
82. Nii-Kono T, Iwasaki Y, Uchida M et al. Indoxyl sulfate induces Endocrinol. Metab. 2014; 99: 3879–88.
skeletal resistance to parathyroid hormone in cultured osteoblastic 99. Riggs BL, Wahner HW, Seeman E et al. Changes in bone mineral
cells. Kidney Int. 2007; 71: 738–43. density of the proximal femur and spine with aging. Differences
83. Barreto FC, Barreto DV, Canziani ME et al. Association between between the postmenopausal and senile osteoporosis syndromes.
indoxyl sulfate and bone histomorphometry in pre-dialysis chronic J. Clin. Invest. 1982; 70: 716–23.
kidney disease patients. J. Bras. Nefrol. 2014; 36: 289–96. 100. Donnelly E, Chen DX, Boskey AL, Baker SP, van der
84. Iwasaki Y, Kazama JJ, Yamato H, Shimoda H, Fukagawa M. Meulen MCH. Contribution of mineral to bone structural behavior
Accumulated uremic toxins attenuate bone mechanical properties and tissue mechanical properties. Calcif. Tissue Int. 2010; 87:
in rats with chronic kidney disease. Bone 2013; 57: 477–83. 450–60.
85. Daya NR, Voskertchian A, Schneider ALC et al. Kidney function 101. Busse B, Bale HA, Zimmermann EA et al. Vitamin D deficiency
and fracture risk: The atherosclerosis risk in communities (ARIC) induces early signs of aging in human bone, increasing the risk of
study. Am. J. Kidney Dis. 2016; 67: 218–26. fracture. Sci. Transl. Med. 2013; 5: 193ra88-93ra88.
86. Mishima T, Motoyama K, Imanishi Y et al. Decreased cortical thickness, 102. Rolvien T, Krause M, Jeschke A et al. Vitamin D regulates
as estimated by a newly developed ultrasound device, as a risk for osteocyte survival and perilacunar remodeling in human and
vertebral fracture in type 2 diabetes mellitus patients with eGFR of murine bone. Bone 2017; 103: 78–87.
less than 60 mL/min/1.73 m2. Osteoporos. Int. 2015; 26: 229–36. 103. Baldock PA, Thomas GP, Hodge JM et al. Vitamin D action and
87. Lima GA, de Paula P-NF, Silva LC et al. Bone density is directly regulation of bone remodeling: Suppression of osteoclastogenesis
associated with glomerular filtration and metabolic acidosis but do by the mature osteoblast. J. Bone Miner. Res. 2006; 21: 1618–26.

94 © 2018 Asian Pacific Society of Nephrology

Вам также может понравиться