You are on page 1of 6

Vitamin D, Proteinuria, Diabetic Nephropathy, and

Progression of CKD
Rajiv Agarwal
Indiana University School of Medicine and Richard L Roudebush VA Medical Center, Indianapolis, Indiana

Although the endocrine effects of vitamin D are widely recognized, somewhat less appreciated is that vitamin D may serve
paracrine functions through local activation by 1-␣-hydroxylase and thus maintain immunity, vascular function, cardiomyo-
cyte health, and abrogate inflammation and insulin resistance. In the kidney, vitamin D may be important for maintaining
podocyte health, preventing epithelial-to-mesenchymal transformation, and suppressing renin gene expression and inflam-
mation. Replacement with pharmacologic dosages of vitamin D receptor agonists (VDRA) in animal models of kidney disease
consistently show reduction in albuminuria, abrogation of glomerulosclerosis, glomerulomegaly, and glomerular inflamma-
tion, effects that may be independent of BP and parathyroid hormone, but the effects of VDRA in preventing tubulointerstitial
fibrosis and preventing the progression of kidney failure in these animal models are less clear. Emerging evidence in patients
with chronic kidney disease (CKD) show that vitamin D can reduce proteinuria or albuminuria even in the presence of
angiotensin-converting enzyme inhibition. In addition to reducing proteinuria, VDRA may reduce insulin resistance, BP, and
inflammation and preserve podocyte loss providing biologic plausibility to the notion that the use of VDRA may be associated
with salubrious outcomes in patients with diabetic nephropathy. Patients with CKD have a very high prevalence of deficiency
of 25-hydroxyvitamin D. Whether pharmacologic dosages of vitamin D instead of VDRA in patients with CKD can overcome
the paracrine and endocrine functions of this vitamin remains unknown. To demonstrate the putative benefits of native
vitamin D and VDRA among patients with CKD, randomized, controlled trials are needed.
Clin J Am Soc Nephrol 4: 1523–1528, 2009. doi: 10.2215/CJN.02010309

I
n 1978, a study published in The Lancet reported 18 pa- patients. Existing data seem to point to a salubrious effect of
tients who had advanced chronic kidney disease (CKD), vitamin D receptor agonists (VDRA) in animals with kidney
creatinine clearance of ⬍35 ml/min, and presence of renal disease, and emerging evidence points to a similar benefit in
osteodystrophy and were treated with either vitamin D3 (4000 patients with CKD. The evolution of science of VDRA in
IU/d) or 1,25-dihydroxyvitamin D [1,25(OH)2D; 1 ␮g/d] along animal models of kidney disease and patients with CKD is
with 500 mg of calcium after a 6-mo observation period (1). In the subject of this review.
the group treated with 1,25(OH)2D, seven of eight patients The effects of vitamin D on the bone and elsewhere can be
developed hypercalcemia that necessitated a reduction in dos- evaluated at the molecular level by examining knockout mice
age. The percentage fall in creatinine clearance was greater and are reviewed by Bouillon et al. (2). Absence of a functional
during treatment than before treatment in all patients who vitamin D receptor (VDR) or its ligand, 25-hydroxyvitamin D-1
were on 1,25(OH)2D (P ⬍ 0.01) and in seven of nine patients on [25(OH)D1] ␣-hydroxylase (CYP27B1), in mice creates a bone
vitamin D3 treatment (although the within group change here and growth plate phenotype that mimics severe vitamin D
was NS). The authors concluded that deterioration of renal deficiency. VDR rescue in the intestine restores a normal bone
function was a major limitation of the clinical use of and growth plate phenotype. Although the VDR is nearly ubiq-
1,25(OH)2D and vitamin D3 in nondialyzed patients with CKD. uitously expressed and almost all cells respond to 1,25(OH)2D
Although the conclusions made by this important clinical exposure, the function of active vitamin D and its receptor is
study were strong, evaluating the state-of-the-art science in not fully overlapping. For example, VDR-deficient mice but not
1978, ⬎30 yr later informs us that the dosage of 1,25(OH)2D vitamin D- or 1 ␣-hydroxylase– deficient mice develop total
was rather high, the sample size too small, and the follow-up alopecia as is also seen in humans. The immune system of
period of 6 mo too short to provide reliable information VDR- or vitamin D– deficient mice shows increased sensitiv-
about the rates of progression of CKD in vitamin D–treated ity to autoimmune diseases such as inflammatory bowel
disease or type 1 diabetes after exposure to predisposing
Published online ahead of print. Publication date available at www.cjasn.org. factors. Although VDR-deficient mice do not have a sponta-
neous increase in cancer, they are more prone to oncogene-
Correspondence: Dr. Rajiv Agarwal, Department of Medicine, Indiana University
School of Medicine, VAMC 111N, 1481 West 10th Street, Indianapolis, IN 46202. or chemocarcinogen-induced tumors. They also develop
Phone: 317-554-0000, ext. 2241; Fax: 317-554-0298; E-mail: ragarwal@iupui.edu high renin hypertension, cardiac hypertrophy, and increased

Copyright © 2009 by the American Society of Nephrology ISSN: 1555-9041/409 –1523


1524 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 4: 1523–1528, 2009

thrombogenicity. Vitamin D deficiency in humans is associ- beneficial cardiovascular effects in patients with CKD, but fur-
ated with increased prevalence of diseases, as predicted by ther research is needed.
the VDR null phenotype.
Vascular Effects
Somjen et al. (9) were the first to report an enzymatically
Animal Studies
active 25(OH)D1 ␣-hydroxylase system in human vascular
Tian et al. (3) reviewed the mechanisms of salutary effects of
smooth muscle cells, which could be upregulated by parathy-
vitamin D on the kidney. The noncalcemic effects of VDRA
roid hormone (PTH) and inhibited by exogenous vitamin D.
include vascular effects, immunomodulatory effects, anti-in-
Emerging experimental data demonstrate important physio-
flammatory effects, suppression of the renin-angiotensin sys-
logic effects of vitamin D on factors that are protective for
tem (RAS), and effects on glucose homeostasis, which are dis-
vascular health (10).
cussed further.

Immunomodulating Effects
Suppression of RAS Vitamin D seems to regulate innate immunity through effects
Seminal discoveries by Li et al. (4) demonstrated that vitamin on microbial recognition peptides such as Toll-like receptors
D is a potent negative endocrine regulator of the RAS and (TLRs) (11). TLRs are membrane-spanning receptors that rec-
works predominantly as a suppressor of renin biosynthesis (5). ognize structurally conserved molecules derived from microbes
Mice that lack the vitamin D receptor have elevated production and activate immune responses. Mycobacterium tuberculosis
of renin and angiotensin II (AngII), leading to hypertension, infection leads to increased 1 ␣-hydroxylase expression and
cardiac hypertrophy, and increased water intake. Vitamin D VDR upregulation in monocyte/macrophages in response to
repression of renin expression is independent of calcium me- activation by TLRs (12). Furthermore, macrophages can also
tabolism, the volume- and salt-sensing mechanisms, and the synthesize antimicrobial peptides such as cathelicidin, an effect
AngII feedback regulation (4). In normal mice, vitamin D defi- that is facilitated in the presence of 25(OH)D (13). In fact, it is
ciency stimulates renin expression, whereas injection of now being recognized that autocrine production of 1,25(OH)2D
1,25(OH)2D reduces renin synthesis (4). In cell cultures, might also be an important negative feedback mechanism to
1,25(OH)2D directly suppresses renin gene transcription by a deactivate innate and inflammatory responses of activated
VDR-dependent mechanism. Combined treatment of diabetic macrophages (14).
mice with losartan and paricalcitol completely prevents albu- In the kidney, vitamin D has similar immunomodulating
minuria, restores glomerular filtration barrier structure, and effects. For example, in vitro, 1,25(OH)2D attenuates TNF-␣–
markedly reduces glomerulosclerosis (6). These beneficial ef- induced monocyte chemoattractant protein 1 (MCP-1) expres-
fects are accompanied by blockade of intrarenal renin and sion by human proximal tubule cells (15). A synthetic VDRA,
AngII accumulation induced by hyperglycemia and losartan. paricalcitol, inhibits renal inflammation by promoting VDR-
These data demonstrate that inhibition of the RAS with com- mediated sequestration of NF-␬B signaling (16). Although cell
bination of vitamin D analogs and RAS inhibitors effectively surface receptor–mediated, nongenomic pathways (17,18) are
prevents renal injury in diabetic nephropathy. 1,25(OH)2D also described, the potent antiproliferative, prodifferentiative, and
suppresses hyperglycemia-induced activation of the RAS and immunomodulating activities (17,19,20) seem to be modulated
TGF-␤ in mesangial and juxtaglomerular cells (7). TGF-␤ acti- via vitamin D receptor– dependent genomic effects; the latter
vates interstitial fibroblasts and induces tubular epithelial-to- seem to provide the biologic basis for salutary effects of VDRA
mesenchymal transition. Thus, by blocking TGF-␤, VDRA have in patients with kidney disease.
the potential to abrogate tubulointerstitial fibrosis. Taken to-
gether, these studies suggest that receptor-mediated vitamin D
Animal Models of Kidney Disease and Benefits of VDRA
actions in part via the RAS may provide renoprotection in Substantial number of studies point out the salutary effects of
diabetic nephropathy. VDRA in animals (Table 1). In aggregate there appears to be a
consistent reduction in albuminuria, abrogation of glomerulo-
Improvement in Insulin Resistance sclerosis, glomerulomegaly and glomerular inflammation, ef-
Vitamin D deficiency has been linked with impaired glucose fects that may be independent of BP and PTH. However, from
metabolism, an established risk factor for cardiovascular dis- these studies the effects of VDRA in preventing tubulointersti-
ease (8). Because vitamin D is often deficient and its metabolism tial fibrosis and preventing the progression of kidney failure
substantially impaired in patients with CKD, absolute or func- are less clear.
tional vitamin D deficiency may be particularly important in
these patients. Furthermore, impaired glucose metabolism is Human Studies
common in CKD. De Boer (8) concluded in a recent review that Vitamin D Deficiency in Patients with CKD
short-term studies on maintenance hemodialysis patients have As renal function declines, serum levels of 1,25(OH)2D pro-
shown that vitamin D treatment improves insulin secretion and gressively decrease, leading to a vitamin D– deficient state (28).
sensitivity, but interventional studies on people without ESRD Despite presence of adequate 25(OH)D, many patients with
have yielded mixed results. Improved glucose metabolism is CKD will have low serum levels of 1,25(OH)2D, which under-
one potential mechanism through which vitamin D may exert lies the functional vitamin D deficiency in these patients. Some-
Table 1. Summary of studiesa
Reference Model Intervention Results

Schwarz et al., Subtotal nephrectomy 1,25(OH)2D Glomerular volume and glomerulosclerosis were less in treated animals.
1998 (21) with and without Albuminuria was 21% that of control animals. No effect on vascular
parathyroidectomy and tubulointerstitial sclerosis, systolic BP, or serum creatinine. Cell
proliferation was less. TGF-␤ expression in vessels and
tubulointerstitium was less. Effects were independent of PTH.
Makibayashi et al., Anti-Thy1 GN 22-Oxa-calcitriol or Less mesangial cell proliferation, less glomerulosclerosis, less
2001 (22) 1,25(OH)2D albuminuria at day 8. Reduced type 1 and IV collagens and ␣-smooth
Clin J Am Soc Nephrol 4: 1523–1528, 2009

muscle actin mRNA expression. TGF-␤ expression in glomeruli


reduced.
Panichi et al., 2001 Anti-Thy1 GN 1,25(OH)2D Proteinuria was 16% at day 7, and urinary IL-6 was significantly less
(23) compared with control animals. Decreased glomerular inflammation
(neutrophil, monocyte), glomerular size, and glomerular cell
proliferation. Creatinine at day 14 not statistically different from
control group.
Hirata et al., Subtotal nephrectomy 22-Oxa-calcitriol Albuminuria less (approximately 25% of controls at 8 wk); prevented
2002 (24) increases in serum creatinine and BUN; and reduced glomerular cell
number, volume, and sclerosis at 8 wk. ␤2-Microglobulin or NAG
levels were not affected.
Kuhlmann et al., Subtotal nephrectomy 1,25(OH)2D Albuminuria less, glomerular volume less, more podocytes, less
2003 (25) podocyte injury, less podocyte hypertrophy.
Tan et al., 2006 Obstructive Paricalcitol Reduced renal interstitial fibrosis, suppressed renal TGF-␤1 and its type
(26) nephropathy I receptor expression, restored vitamin D receptor abundance, and
inhibited cell proliferation and apoptosis. Paricalcitol also blocked
directly the EMT.
Freundlich et al., Subtotal nephrectomy Paricalcitol Glomerular and tubulointerstitial damage, hypertension, proteinuria,
2008 (27) and the deterioration of renal function were improved as a result of
decrease in angiotensinogen, renin, renin receptor, and vascular
endothelial growth factor mRNA levels in the remnant kidney by 30
to 50% with paricalcitol treatment.
Zhang et al., Diabetic VDR 1,25(OH)2D Diabetic VDR knockout mice developed more severe albuminuria and
2008 (7) knockout mice glomerulosclerosis as a result of increased glomerular basement
membrane thickening and podocyte effacement. In receptor knockout
mice, increased renin, angiotensinogen, TGF-␤, and connective tissue
growth factor accompanied the more severe renal injury.
Moving Points in Nephrology

a
1,25(OH)2D, 1,25-dihydroxyvitamin D; BUN, blood urea nitrogen; EMT, epithelial-to-mesenchymal transition; GN, glomerulonephritis; PTH, parathyroid hormone;
VDR, vitamin D receptor.
1525
1526 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 4: 1523–1528, 2009

what less commonly recognized is the high prevalence of nu- reported that 25(OH)D as well as its active metabolite
tritional vitamin D deficiency, as determined by low 25(OH)D 1,25(OH)2D is directly associated with endothelial function and
concentrations in these patients. Thus, “absolute” vitamin D inversely with arterial calcifications in patients with CKD.
deficiency is also common in patients with CKD. In 43 patients Taken together, it seems that 25(OH)D deficiency in patients
with CKD spanning an estimated GFR (eGFR) range of 11 to with CKD is associated with worse risk factors (albuminuria,
111 ml/min per 1.73 m2, Gonzalez et al. (29) reported vitamin D insulin resistance, inflammation, hypertension, dyslipidemia,
deficiency as defined by a serum 25(OH)D level ⬍30 ng/ml in and endothelial dysfunction) for progression of kidney disease.
86% of the patients. Possible explanations for this high preva- The presence of vitamin D deficiency is associated with in-
lence of nutritional D deficiency in patients are reduced sun- cident cardiovascular disease in the general population (36).
light exposure (e.g., in the elderly, the sick, dark-skinned peo- Because risk factors for cardiovascular and kidney disease by
ple, those who wear a veil for cultural reasons), losses of and large overlap, it is possible that cardiovascular and renal
vitamin D– binding protein in proteinuric states [25(OH)D is benefits may accrue in patients with CKD. In fact, several
lost in the urine bound to vitamin D– binding protein] and a studies of patients who had CKD and were not on dialysis
compromised endogenous synthesis of vitamin D from the skin supported the presence of a salutary effect of vitamin D. In an
in the uremic state. It seems that increased removal of 25(OH)D Italian cohort, Ravani et al. (37) found that 25(OH)D plasma
via nonrenal pathways may also play a role in causing a low concentration was an independent inverse predictor of disease
25(OH)D level. In fact, it is believed that the vast majority of progression and death in patients with stages 2 through 5 CKD
25(OH)D is used by the nonrenal tissues. Because monocytes after multivariate adjustment. Shoben et al. (38) reported that
and macrophages express 1-␣ hydroxylase, it seems plausible oral calcitriol use was associated with lower mortality in non-
that apparent nutritional vitamin D deficiency could be a man- dialysis US veterans with CKD. After multivariate adjustment
ifestation of the inflammatory state in patients with CKD. In including eGFR and baseline levels of PTH, calcium, and phos-
this context, VDRA administration may lead to reduction in the phorous, oral calcitriol use was associated with a 26% lower
inflammatory state (30). Whether similar benefits can be real- risk for death and a 20% lower risk for death or dialysis. The
ized with vitamin D administration needs to be studied in association of calcitriol with improved survival was not statis-
future trials. Endothelial cells can also rapidly induce 1-␣ hy- tically different across baseline PTH levels, although calcitriol
droxylase activity in response to inflammatory cytokines and use was associated with a greater risk for hypercalcemia. In
may serve to deplete the stores of 25(OH)D (31). In one small another cohort of US veterans with CKD, Kovesdy et al. (39)
study, VDRA was unable to improve endothelial function, but reported that the incidence rate ratios for mortality and com-
a larger study with vitamin D is warranted. bined death and dialysis initiation were significantly lower in
patients who were treated with calcitriol. Treatment with cal-
Vitamin D and Cohort Studies in CKD citriol was also associated with a trend toward a lower inci-
In 15,068 adults who participated in the Third National dence of dialysis.
Health and Nutrition Examination Survey (NHANES III), de It remains that neither cross-sectional nor longitudinal stud-
Boer et al. (32) reported a stepwise increase in the prevalence of ies can prove a cause-and-effect relationship between use of
albuminuria with decreasing quartiles of 25(OH)D concentra- VDRA and outcomes. Randomized trials are needed.
tion. After multivariate adjustment including region and season
of measurement and eGFR, relative risks for albuminuria by Randomized Trials of VDRA in Patients with CKD
decreasing quartile of vitamin D concentration were signifi- At least three clinical trials have evaluated the use of VDRA
cantly increased to 1.14, 1.22, and 1.37 in successive quartiles. In in patients who had CKD and were not on renal replacement
the NHANES survey, 25(OH)D deficiency and eGFR both were therapy. In the first study, Agarwal et al. (40) reported reduc-
independently related to insulin resistance (33) as well as sev- tion in proteinuria detected semiquantitatively by dipstick us-
eral other cardiovascular risk factors, such as hypertension, ing an automated analysis in patients who had stages 3 and 4
diabetes, obesity, and high serum triglyceride levels (34). CKD with secondary hyperparathyroidism and participated in
Zehnder et al. (15) measured vitamin D metabolites, markers of three randomized, controlled trials of oral paricalcitol. In that
inflammation and gene expression, in 174 patients with a vari- post hoc analysis, reduction in dipstick proteinuria occurred in
ety of kidney diseases. Urinary MCP-1 and renal macrophage the face of the frequent use of agents that block the RAS.
infiltration each were inversely correlated with serum Although previous studies reported that dipstick proteinuria is
1,25(OH)2D levels. Higher levels of 25(OH)D were also associ- a good predictor of spot urine protein/creatinine ratios, an
ated with lower inflammation. Patients with acute renal inflam- obvious limitation of that study was the method of detection of
mation had a significant increase in urinary and kidney MCP-1, proteinuria (automated dipstick versus spot urinary protein/
macrophage infiltration, and macrophage and renal epithelial creatinine or albumin/creatinine ratio).
1-␣ hydroxylase (CYP27B1) expression but significantly lower Extending these findings, Alborzi et al. (30) reported a pro-
levels of serum 1,25(OH)2D in comparison with patients with spective pilot trial of 24 patients who had CKD (two patients
chronic ischemic disease despite similar levels of renal damage. stage 2, remaining stage 3) and were randomly allocated in a
Thus, decreased serum vitamin D metabolites and activation of double-blind manner to three equal groups to receive 0, 1, or 2
the paracrine/autocrine vitamin D system are important corre- ␮g of paricalcitol, a vitamin D analog, orally for 1 mo. At 1 mo,
lates of inflammation in patients with CKD. London et al. (35) treatment/baseline ratio of high-sensitivity C-reactive protein
Clin J Am Soc Nephrol 4: 1523–1528, 2009 Moving Points in Nephrology 1527

was 1.5 (95% confidence interval [CI] 1.1 to 2.1; P ⫽ 0.02) with per 1.73 m2 and urine albumin/creatinine concentration be-
placebo, 0.8 (95% CI 0.3 to 1.9; P ⫽ 0.62) with 1-␮g dosage, and tween 100 and 3000 mg/d on average of three morning void
0.5 (95% CI 0.3 to 0.9; P ⫽ 0. 03) with 2-␮g dosage of paricalcitol. specimen. All patients have to be on a stable dosage of angio-
At 1 mo, treatment/baseline ratio of 24-h albumin excretion tensin-converting enzyme inhibitor or angiotensin receptor
rate was 1.35 (95% CI 1.08 to 1.69; P ⫽ 0.01) with placebo, 0.52 blocker and have reasonable control of BP and blood glucose to
(95% CI 0.40 to 0.69; P ⬍ 0.001) with 1-␮g dosage, and 0.54 (95% participate. Results are awaited. Each of these trials, conducted
CI 0.35 to 0.83; P ⫽ 0. 01) with 2-␮g dosage (P ⬍ 0.001 for with patients with CKD, should provide better evidence (or
between-group changes). No differences were observed in lack thereof) for cardiovascular and renal protection.
iothalamate clearance, flow-mediated dilation (a measure of
endothelial function), 24-h ambulatory BP, or PTH with treat- Disclosures
ment or upon washout. The limitations of that trial include the Consultant, speaker bureau, and grant support from Abbott.
small sample size and the limited duration of exposure to VDR
activator. Furthermore, all patients had low 25(OH)D defi-
ciency in that study; whether nutritional replacement could References
have improved albuminuria or inflammation remains un- 1. Christiansen C, Rodbro P, Christensen MS, Hartnack B,
known. That study therefore cannot provide definite answers Transbol I: Deterioration of renal function during treat-
to questions of the clinical use of paricalcitol for renal protec- ment of chronic renal failure with 1,25-dihydroxycholecal-
tion; however, it seems from that study that paricalcitol-in- ciferol. Lancet 2: 700 –703, 1978
2. Bouillon R, Carmeliet G, Verlinden L, van EE, Verstuyf A,
duced reduction in albuminuria and inflammation may be
Luderer HF, Lieben L, Mathieu C, Demay M: Vitamin D
mediated independent of its effects on hemodynamics or PTH
and human health: Lessons from vitamin D receptor null
suppression. mice. Endocr Rev 29: 726 –776, 2008
In an uncontrolled trial, reported from Hong Kong, 10 pa- 3. Tian J, Liu Y, Williams LA, de Zeeuw D: Potential role of
tients with biopsy-proven IgA nephropathy and persistent pro- active vitamin D in retarding the progression of chronic
teinuria despite angiotensin-converting enzyme inhibition and kidney disease. Nephrol Dial Transplant 22: 321–328, 2007
AngII receptor blockade were treated with 0.5 ␮g calcitriol 4. Li YC, Kong J, Wei M, Chen ZF, Liu SQ, Cao LP: 1,25-
twice weekly for 12 wk (41). After calcitriol treatment, there Dihydroxyvitamin D(3) is a negative endocrine regulator
was a significant overall decrease in urine protein-creatinine of the renin-angiotensin system. J Clin Invest 110: 229 –238,
ratio from 1.98 ⫾ 0.74 to 1.48 ⫾ 0.81 g/g (P ⫽ 0.007) during the 2002
first 6 wk that persisted throughout the study period. No 5. Li YC, Qiao G, Uskokovic M, Xiang W, Zheng W, Kong J:
Vitamin D: A negative endocrine regulator of the renin-
significant change in BP or renal function was noted. There was
angiotensin system and blood pressure. J Steroid Biochem
a simultaneous decrease in serum TGF-␤ level, and percentage
Mol Biol 89 –90: 387–392, 2004
of decrease in serum TGF-␤ level significantly correlated with 6. Zhang Z, Zhang Y, Ning G, Deb DK, Kong J, Li YC:
percentage of change in proteinuria (Spearman r ⫽ 0.643; P ⫽ Combination therapy with AT1 blocker and vitamin D
0.02). analog markedly ameliorates diabetic nephropathy: Block-
Whether VDRA should be used to reduce the rate of progres- ade of compensatory renin increase. Proc Natl Acad Sci
sion of CKD cannot be answered by the studies discussed U S A 105: 15896 –15901, 2008
herein; larger trials are required. Lowering of albuminuria or 7. Zhang Z, Sun L, Wang Y, Ning G, Minto AW, Kong J,
proteinuria in these trials, however, occurred without changes Quigg RJ, Li YC: Renoprotective role of the vitamin D
in BP even when recorded by ambulatory BP monitoring, rais- receptor in diabetic nephropathy. Kidney Int 73: 163–171,
ing the notion that improvement in kidney disease conferred by 2008
8. De Boer I: Vitamin D and glucose metabolism in chronic
the use of these drugs may occur via nonhemodynamic path-
kidney disease. Curr Opin Nephrol Hypertens 17: 566 –572,
ways.
2008
9. Somjen D, Weisman Y, Kohen F, Gayer B, Limor R, Sharon
Future Directions O, Jaccard N, Knoll E, Stern N: 25-Hydroxyvitamin D3–
Randomized, controlled trials with paricalcitol are under 1alpha-hydroxylase is expressed in human vascular
way to demonstrate whether paricalcitol will have antipro- smooth muscle cells and is upregulated by parathyroid
teinuric or cardioprotective effects in patients with CKD. In hormone and estrogenic compounds. Circulation 111:
one such trial, reduction in albuminuria from baseline to 24 1666 –1671, 2005
wk is the primary end point (ClinicalTrials.gov identifier 10. Zittermann A, Koerfer R: Protective and toxic effects of
NCT00421733), whereas, in another, the change in left ventric- vitamin D on vascular calcification: Clinical implications.
Mol Aspects Med 29: 423– 432, 2008
ular mass index assessed by cardiac magnetic resonance imag-
11. Maalouf NM: The noncalciotropic actions of vitamin D:
ing over 48 wk is the primary end point (ClinicalTrials.gov
Recent clinical developments. Curr Opin Nephrol Hypertens
identifier NCT00497146). The Vitamin D Receptor Activator 17: 408 – 415, 2008
(Paricalcitol) in Albuminuria Lowering (VITAL) study has ran- 12. Liu PT, Stenger S, Li H, Wenzel L, Tan BH, Krutzik SR,
domly assigned 282 patients with diabetes in a double-blind, Ochoa MT, Schauber J, Wu K, Meinken C, Kamen DL,
multicenter trial to placebo or 1 or 2 ␮g of paricalcitol taken Wagner M, Bals R, Steinmeyer A, Zugel U, Gallo RL,
once daily in patients with eGFR between 15 and 90 ml/min Eisenberg D, Hewison M, Hollis BW, Adams JS, Bloom BR,
1528 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 4: 1523–1528, 2009

Modlin RL: Toll-like receptor triggering of a vitamin D- 28. Levin A, Bakris GL, Molitch M, Smulders M, Tian J, Wil-
mediated human antimicrobial response. Science 311: liams LA, Andress DL: Prevalence of abnormal serum
1770 –1773, 2006 vitamin D, PTH, calcium, and phosphorus in patients with
13. Selvaraj P, Prabhu AS, Harishankar M, Alagarasu K: chronic kidney disease: Results of the study to evaluate
Plasma 1,25-dihydroxy vitamin D(3) level and expression early kidney disease. Kidney Int 71: 31–38, 2006
of vitamin d receptor and cathelicidin in pulmonary tuber- 29. Gonzalez EA, Sachdeva A, Oliver DA, Martin KJ: Vitamin
culosis. J Clin Immunol February 14, 2009 [epub ahead of D insufficiency and deficiency in chronic kidney disease: A
print] single center observational study. Am J Nephrol 24: 503–510,
14. Helming L, Bose J, Ehrchen J, Schiebe S, Frahm T, Geffers 2004
R, Probst-Kepper M, Balling R, Lengeling A: 1Alpha,25- 30. Alborzi P, Patel NA, Peterson C, Bills JE, Bekele DM,
dihydroxyvitamin D3 is a potent suppressor of interferon Bunaye Z, Light RP, Agarwal R: Paricalcitol reduces albu-
gamma-mediated macrophage activation. Blood 106: 4351– minuria and inflammation in chronic kidney disease: A
4358, 2005 randomized double-blind pilot trial. Hypertension 52: 249 –
15. Zehnder D, Quinkler M, Eardley KS, Bland R, Lepenies J, 255, 2008
Hughes SV, Raymond NT, Howie AJ, Cockwell P, Stewart 31. Zehnder D, Bland R, Chana RS, Wheeler DC, Howie AJ,
PM, Hewison M: Reduction of the vitamin D hormonal Williams MC, Stewart PM, Hewison M: Synthesis of 1,25-
system in kidney disease is associated with increased renal dihydroxyvitamin D(3) by human endothelial cells is reg-
inflammation. Kidney Int 74: 1343–1353, 2008 ulated by inflammatory cytokines: A novel autocrine de-
16. Tan X, Wen X, Liu Y: Paricalcitol inhibits renal inflamma- terminant of vascular cell adhesion. J Am Soc Nephrol 13:
tion by promoting vitamin D receptor-mediated sequestra- 621– 629, 2002
tion of NF-kappaB signaling. J Am Soc Nephrol 19: 1741– 32. De Boer I, Ioannou GN, Kestenbaum B, Brunzell JD, Weiss
1752, 2008 NS: 25-Hydroxyvitamin D levels and albuminuria in the
17. Seibert E, Levin NW, Kuhlmann MK: Immunomodulating Third National Health and Nutrition Examination Survey
effects of vitamin D analogs in hemodialysis patients. He- (NHANES III). Am J Kidney Dis 50: 69 –77, 2007
modial Int 9[Suppl 1]: S25–S29, 2005 33. Chonchol M, Scragg R: 25-Hydroxyvitamin D, insulin re-
18. Dusso AS, Brown AJ, Slatopolsky E: Vitamin D. Am J
sistance, and kidney function in the Third National Health
Physiol Renal Physiol 289: F8 –F28, 2005
and Nutrition Examination Survey. Kidney Int 71: 134 –139,
19. Mathieu C, Adorini L: The coming of age of 1,25-dihy-
2007
droxyvitamin D(3) analogs as immunomodulatory agents.
34. Martins D, Wolf M, Pan D, Zadshir A, Tareen N, Thadhani
Trends Mol Med 8: 174 –179, 2002
R, Felsenfeld A, Levine B, Mehrotra R, Norris K: Preva-
20. Lemire JM: Immunomodulatory role of 1,25-dihydroxyvi-
lence of cardiovascular risk factors and the serum levels of
tamin D3. J Cell Biochem 49: 26 –31, 1992
25-hydroxyvitamin D in the United States: Data from the
21. Schwarz U, Amann K, Orth SR, Simonaviciene A, Wessels
Third National Health and Nutrition Examination Survey.
S, Ritz E: Effect of 1,25 (OH)2 vitamin D3 on glomerulo-
Arch Intern Med 167: 1159 –1165, 2007
sclerosis in subtotally nephrectomized rats. Kidney Int 53:
35. London GM, Guerin AP, Verbeke FH, Pannier B, Boutouy-
1696 –1705, 1998
rie P, Marchais SJ, Metivier F: Mineral metabolism and
22. Makibayashi K, Tatematsu M, Hirata M, Fukushima N,
Kusano K, Ohashi S, Abe H, Kuze K, Fukatsu A, Kita T, arterial functions in end-stage renal disease: Potential role
Doi T: A vitamin D analog ameliorates glomerular injury of 25-hydroxyvitamin D deficiency. J Am Soc Nephrol 18:
on rat glomerulonephritis. Am J Pathol 158: 1733–1741, 2001 613– 620, 2007
23. Panichi V, Migliori M, Taccola D, Filippi C, De Nisco L, 36. Wang TJ, Pencina MJ, Booth SL, Jacques PF, Ingelsson E,
Giovannini L, Palla R, Tetta C, Camussi G: Effects of Lanier K, Benjamin EJ, D’Agostino RB, Wolf M, Vasan RS:
1,25(OH)2D3 in experimental mesangial proliferative ne- Vitamin D deficiency and risk of cardiovascular disease.
phritis in rats. Kidney Int 60: 87–95, 2001 Circulation 117: 503–511, 2008
24. Hirata M, Makibayashi K, Katsumata K, Kusano K, Wa- 37. Ravani P, Malberti F, Tripepi G, Pecchini P, Cutrupi S,
tanabe T, Fukushima N, Doi T: 22-Oxacalcitriol prevents Pizzini P, Mallamaci F, Zoccali C: Vitamin D levels and
progressive glomerulosclerosis without adversely affecting patient outcome in chronic kidney disease. Kidney Int 75:
calcium and phosphorus metabolism in subtotally ne- 88 –95, 2009
phrectomized rats. Nephrol Dial Transplant 17: 2132–2137, 38. Shoben AB, Rudser KD, de Boer, I, Young B, Kestenbaum
2002 B: Association of oral calcitriol with improved survival in
25. Kuhlmann A, Haas CS, Gross ML, Reulbach U, Holzinger nondialyzed CKD. J Am Soc Nephrol 19: 1613–1619, 2008
M, Schwarz U, Ritz E, Amann K: 1,25-Dihydroxyvitamin 39. Kovesdy CP, Ahmadzadeh S, Anderson JE, Kalantar-Za-
D3 decreases podocyte loss and podocyte hypertrophy in deh K: Association of activated vitamin D treatment and
the subtotally nephrectomized rat. Am J Physiol Renal mortality in chronic kidney disease. Arch Intern Med 168:
Physiol 286: F526 –F533, 2004 397– 403, 2008
26. Tan X, Li Y, Liu Y: Paricalcitol attenuates renal interstitial 40. Agarwal R, Panesar A, Lewis RR: Dipstick proteinuria:
fibrosis in obstructive nephropathy. J Am Soc Nephrol 17: Can it guide hypertension management? Am J Kidney Dis
3382–3393, 2006 39: 1190 –1195, 2002
27. Freundlich M, Quiroz Y, Zhang Z, Zhang Y, Bravo Y, 41. Szeto CC, Chow KM, Kwan BC, Chung KY, Leung CB, Li
Weisinger JR, Li YC, Rodriguez-Iturbe B: Suppression of PK: Oral calcitriol for the treatment of persistent protein-
renin-angiotensin gene expression in the kidney by pari- uria in immunoglobulin A nephropathy: An uncontrolled
calcitol. Kidney Int 74: 1394 –1402, 2008 trial. Am J Kidney Dis 51: 724 –731, 2008

You might also like