Acessibilidade / Reportar erro

Vascular calcification in chronic kidney disease: a review

Abstracts

Vascular calcification (VC), an independent and strong predictor of cardiovascular risk, is often found in CKD patients. The degree of VC is providing incremental prognostic value over traditional risk markers. There is interest in improving our understanding of mechanisms, establishing diagnostic methods and effective prevention and treatment modalities. The abnormal mineral metabolism of CKD is known to facilitate the progression of VC, in concert with altered activities of VC inhibitors. Possible measures to prevent VC include the control of serum calcium and phosphate as well as other factors involved in its progression, including vitamin D sterols, parathyroid hormone, fibroblast growth factor-23, klotho, and VC inhibitors. In addition, we discuss new possible therapeutic approaches to halt VC or reverse its progression. The principal aim of this review is to provide an updated overview of VC in patients with CKD, with particular focus on pathophysiology, diagnosis, prevention and treatment.

chronic, renal insufficiency; review; vascular calcification


Pacientes com doença renal crônica (DRC) frequentemente apresentam calcificação vascular (CV) - um forte e independente fator preditor de risco cardiovascular. O grau da CV tem proporcionado maior valor prognóstico quando comparado a outros marcadores mais tradicionais de risco. Há muito interesse em aprimorar nosso conhecimento sobre os mecanismos, estabelecer métodos diagnósticos e desenvolver modalidades mais eficazes de prevenção e tratamento. Sabe-se que a anormalidade metabólica encontrada na DRC facilita a progressão da CV juntamente com alterações nas atividades dos inibidores da CV. Possíveis medidas para se evitar a CV incluem o controle do cálcio e fosfato séricos, assim como outros fatores envolvidos em sua progressão, incluindo ésteres da vitamina D, hormônio da paratireoide, fator 23 de crescimento de fibroblastos, klotho e inibidores da CV. Além disso, discutimos novas abordagens terapêuticas para interromper a progressão da CV e reverter sua ação. O principal objetivo dessa revisão é proporcionar uma atualização sobre a CV em pacientes com DRC, concentrando-se mais especificamente em sua fisiopatologia, diagnóstico, prevenção e tratamento.

calcificação vascular; insuficiência renal, crônica; revisão


UPDATE ARTICLE

Vascular calcification in chronic kidney disease: a review

Rodrigo Bueno de OliveiraI; Hirokazu OkazakiII; Andrea E. Marques StinghenIII; Tilman B. DrüekeIV; Ziad A. MassyV; Vanda JorgettiVI

IINSERM U-1088, UFR de Médecine et de Pharmacie, Université de Picardie Jules Verne, Amiens, France and Division of Nephrology, University of São Paulo, São Paulo, Brazil

IIINSERM U-1088, UFR de Médecine et de Pharmacie, Université de Picardie Jules Verne, Amiens, France and Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University, Graduate School of Medicine, Osaka, Japan

IIIINSERM U-1088, UFR de Médecine et de Pharmacie, Université de Picardie Jules Verne, Amiens, France and Department of Basic Pathology, Experimental Nephrology Laboratory, Universidade Federal do Paraná, Curitiba, Brazil

IVINSERM U-1088, UFR de Médecine et de Pharmacie, Université de Picardie Jules Verne, Amiens, France

VDivision of Nephrology, Ambroise Paré Hospital, Paris Ile de France Ouest (UVSQ) University, Boulogne Billancourt/Paris, and INSERM U-1088, Université de Picardie Jules Verne, Amiens, France

VIDivision of Nephrology, University of São Paulo, São Paulo, Brazil

Correspondence to Correspondence to: Vanda Jorgetti Division of Nephrology, University of São Paulo, São Paulo, Brazil Av. Dr. Arnaldo, nº 455, 3º andar, sala 334 São Paulo, SP, Brazil. CEP: 01246-903 Tel: +55 (11) 3061-8351

ABSTRACT

Vascular calcification (VC), an independent and strong predictor of cardiovascular risk, is often found in CKD patients. The degree of VC is providing incremental prognostic value over traditional risk markers. There is interest in improving our understanding of mechanisms, establishing diagnostic methods and effective prevention and treatment modalities. The abnormal mineral metabolism of CKD is known to facilitate the progression of VC, in concert with altered activities of VC inhibitors. Possible measures to prevent VC include the control of serum calcium and phosphate as well as other factors involved in its progression, including vitamin D sterols, parathyroid hormone, fibroblast growth factor-23, klotho, and VC inhibitors. In addition, we discuss new possible therapeutic approaches to halt VC or reverse its progression. The principal aim of this review is to provide an updated overview of VC in patients with CKD, with particular focus on pathophysiology, diagnosis, prevention and treatment.

Keywords: chronic, renal insufficiency; review; vascular calcification.

INTRODUCTION

Based on the Health Science Descriptors definition,1 vascular calcification (VC) is a pathologic process characterized by thickening and loss of elasticity of the muscular arteries walls due to calcification of the tunica media and/or intima.

The majority of authors refer to VC as a general term for both intima and media calcification. However, there are differences between them. Intima calcification, which typically occurs within atherosclerotic plaque in the aorta, coronary arteries, and other large arteries, is an indicator of advanced stages of atherosclerosis. Media calcification, which is often found in patients with metabolic syndrome, diabetes and/or chronic kidney disease (CKD), is characterized by diffuse mineral deposition along elastic fibers in both elastic-type conduit arteries and muscle-type resistance arteries.

VC is a common problem among CKD patients. Its prevalence increases with the progressive decrease of kidney function.2 The association between soft-tissue calcification and uremia has been recognized more than 100 years ago. At that time, Rudolf Virchow (1821-1902) described the presence of extraosseous calcifications in a group of patients with non-metabolic bone disease, uremia, primary hyperparathyroidism and vitamin D intoxication. Virchow proposed an interesting hypothesis to explain this phenomenon: he supposed that calcium salts were dissolved from bone and carried by the blood stream to be deposited at some distant site to form "calcium metastases", a process analogous to the dissemination of malignant cells from a primary neoplasm.3 Contemporaneously, Karl Rokitansky (1804-1878) made another important contribution by describing the presence of atherosclerotic lesions.4 Some years later, in 1903, Johann Georg Mönckeberg described the medial type of VC which he named calcific sclerosis.5

Although the first descriptions of media calcification were made a long time ago, it became a widely recognized problem only after the introduction of long-term dialysis and renal transplantation. In the past decades, studies in CKD patients showed that the degree of VC was an independent and strong predictor of death in association with vessel stiffness, arterial hypertension, left ventricular hypertrophy and cardiomyopathy.6,7

Therefore, great efforts have been made to improve our understanding of the pathogenesis, diagnosis, prevention and treatment of VC. The purpose of this review is to provide an update of our present insight into the VC related to CKD.

PATHOGENESIS

GENERAL MECHANISMS OF VASCULAR CALCIFICATION

VC is a complex process, involving not only simple precipitation of supersaturated phosphate and calcium concentrations in the extracellular milieu (mineral step) but also a tightly regulated, cell mediated process including apoptosis, osteochondrogenic differentiation, and elastin degradation (cellular step). The time course of these two steps in vivo, in particular to know which one appears first, remains poorly defined. The main pathogenetic events are summarized below.

APOPTOSIS

Vascular smooth muscle cells (VSMC) apoptosis is regarded as an important contributor in the initiation of VC. Of note, VSMC within atheromatous plaques exhibit increased sensitivity to apoptosis compared with those in normal vessel wall.8,9 Apoptotic bodies derived from VSMC are thought to play a role as nucleating structures for calcium crystal formation such as matrix vesicles in the initiation of calcification.10,11

OSTEOCHONDROGENIC DIFFERENTIATION

VSCM phenotype change to osteochondrogenic cells is characterized by the appearance of matrix vesicles containing apatite and calcifying collagen fibrils on the surface of VSMC. As in bone, these vesicles act as early nucleation sites for calcification. Moreover, VSMC synthesize bony-associated proteins and promote crystal formation and deposition.12In vitro studies have demonstrated this phenotypic change, featured by the expression of bone-associated proteins including alkaline phosphatase (ALP), osteocalcin, and osteopontin. Runt-related transcription factor 2 (Runx2) and Msh homeobox 2, which are obligate transcription factors in normal bone development, also have been shown to be associated with VSMC osteochondrogenic differentiation.13

The phenotypic change of VSMC has also been demonstrated in in vivo studies. Apolipoprotein-E and matrix Gla protein (MGP) knockout mice exhibit osteochondrocyte-like cells flanking the calcium deposits in the vessel wall.14,15 In human calcified arteries, the expression of collagen II and sex determining region Y related high-mobility group box 9 (sox9), which are keys transcription factors for chondrogenesis, has been documented as well.16

The VSMC nature of osteochondroblast-like cells in VC has recently been questioned by Tang Z et al.17 These authors claimed that in response to vascular injuries, multipotent vascular stem cells, not mature VSMC, differentiate into osteochondrogenic cells and thereby initiate VC. This hypothesis has been immediately refuted by Nguyen et al.18 who stated that there is compelling evidence that VSMC are not terminally differentiated and are capable of transition into a phenotype characterized by cell proliferation and loss of differentiation markers.

ELASTIN DEGRADATION

Elastic lamellae consist mainly of amorphous elastin, the major component of the aortic medial layer, in addition to the concentric layers of helically arranged smooth muscle cells. Elastocalcinosis is characterized by deposition of hydroxyapatite on the elastic lamellae of the arteries. Elastin degradation is known to play an important role in the initiation and progression of VC. It is induced by elastase, metalloproteinases and other proteases including cysteine and serine. Degraded elastin has a high affinity for calcium, facilitating growth of hydroxyapatite along the elastic lamellae. Moreover, elastin derived peptides binding to elastin laminin receptors on the surface of VSMC and through transforming growth factor-β signaling can increase proliferation and upregulate Runx-2, resulting in osteochondrogenic differentiation.19,20

MOLECULAR MECHANISMS OF VASCULAR CALCIFICATION

In addition to the mechanisms described above, it has become increasingly clear that there are several inhibitory proteins and promoters involved in the process of VC (Table 1).21-25 A complex interplay between promoters including bone morphogenic protein 2 (BMP-2) and receptor activator of nuclear factor-kappa B ligand (RANKL), and inhibitors including MGP, BMP-7, osteoprotegerin, fetuin-A, and osteopontin, regulates this process. Actually, it has also been speculated that promoter molecules can act via microRNA (miR) regulation. Recently, Balderman et al.26 shown that human VSMC treated by BMP-2 downregulate miR-30b and miR-30c to increase Runx2 expression and promote mineralization.

The following paragraphs describe the main molecular mechanisms involved in VC.

PHOSPHATE METABOLIC PATHWAY

Phosphate homeostasis is maintained by the hormonal control of its transport in intestine, bone, and kidney. The most active form of vitamin D, 1,25-dihydroxyvitamin D [1,25 diOH D], which is synthesized in the kidney, increases the intestinal absorptionofphosphateandstimulatesosteoclastogenesis in bone, leading to an increase in extracellular phosphate concentration. PTH acts on the kidney to stimulate both 1,25 diOH D synthesis through activation of 1α-25OH D hydroxylase, and urinary phosphate excretion.27 In addition to PTH and 1,25 diOH D, FGF23 and Klotho have been discovered more recently as novel factors involved in phosphate metabolism. The importance of the Klotho - FGF23 axis will be discussed below.

Phosphate is a well-known inducer of VSMC apoptosis and osteochondrogenic differentiation. Increased phosphate levels suppress both the expression of growth arrest specific gene 6 (Gas6) and its receptor in VSMC.28 This inhibition leads to the suppression of phosphatidyl inositol-3 kinase PI3K/Akt survival pathway, and thereby favors VSMC apoptosis.29

Phosphate transport into the cell is primarily mediated by sodium-dependent phosphate (Na/Pi) cotransporters. Its uptake into VSMC occurs mainly via phosphate transporter 1 (Pit-1), a member of the type III Na/Pi co-transporters, leading to osteochondrogenic differentiation.30 Of note, Pit-1 knockdown has been shown to suppress phosphate-induced calcification and to block induction of Runx2 and osteopontin.31

PYROPHOSPHATE (PPI)

PPi is a major physiological inhibitor of hydroxyapatite formation. It also potently inhibits VC. PPi is generated by hydrolysis of ATP induced by the enzyme ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (Enpp1), an extracellular membrane-bound glycoprotein. When the Enpp1 gene is mutated and the enzyme is inactive, like in the rare clinical syndrome called "idiopathic infantile arterial calcification", severe calcification of the internal elastic lamina of muscle arteries ensues. This represents one among the numerous demonstrations of the important role of PPi in preventing VC.32 In contrast, ALP, whose expression is induced by VSMC osteochondrogenic transformation, promotes vessel wall matrix mineralization. One of the main roles of ALP is to hydrolyze PPi, which in turn generates phosphate ions and thereby favors the development of VC.

KLOTHO - FIBROBLAST GROWTH FACTOR-23 AXIS

In addition to its well-known anti-aging action, Klotho serves as a co-factor of FGF23 in conferring FGF23 specificity for FGF receptor activation. The two together play an essential role in the control of phosphate and vitamin D metabolism, by enhancing urinary phosphate excretion and suppressing renal 1α-25OH vitamin D hydroxylase activity. Klothodeficient mice and FGF23-null mice exhibit a similar phenotype, characterized by an accelerated aging process with shortened life span, atherosclerosis, and soft-tissue calcification including VC.33,34 Interestingly, Klotho overexpression in CKD mice strikingly reduces aortic calcification, as compared to Klotho-deficient and wild-type mice with similar renal function impairment. Klotho also is capable of directly inhibiting phosphate-induced calcification of VSMC in vitro.35 Regarding FGF-23, its role in VC is less clear in the context of CKD. As an apparent paradox, high serum FGF23 were also found to be associated with the severity of atherosclerosis36 and VC in patients with CKD.37 Curiously, other study involving non-CKD patients observed a negative association between FGF-23 and the presence of atherosclerotic lesion.38 Several recent studies, but not all, showed an involvement of both Klotho and FGF23 in VC, both in interaction and separately from each other, in addition to calcium, phosphate, PTH and 1,25 diOH D.39,40 However, their exact and respective roles in this process require further study.

CALCIUM-SENSING RECEPTOR

The CaR is a G protein-coupled receptor. It is expressed in tissues which are involved in the regulation of calcium metabolism, including the parathyroid, thyroid, kidney, bone and intestine.41 In addition, the CaR has also shown to be expressed in blood vessels, both in endothelial cells42 and VSMC.43 Alam et al.44 showed that a reduction in CaR expression in VSMC is associated with increased mineralization, and that calcimimetics can attenuate mineral deposition. Ivanovski et al.45 showed a protective effect of calcimimetics against the progression of VC and atherosclerosis in uremic apolipoprotein-E knockout mice. They also showed a direct inhibition by calcimimetics of phosphate-induced human VSMC calcification in vitro. This inhibitory effect could be abolished by suppression of CaR expression. Koleganova et al.46 reported that calcimimetics increase MGP expression and decrease Pit1 expression in uremic rats, preventing vessel wall remodeling. These observations suggest that calcimimetics play an important role in VC not only by indirect effects, such as improved control of serum PTH, calcium and phosphate levels, but also by a direct effect on vascular cells. These findings underline the importance of research into the role of CaR in the pathogenesis of of VC.

OXIDATIVE STRESS

Oxidative stress can be defined as a disturbance of normal cellular and molecular function caused by an imbalance between the excessive production of reactive oxygen species (ROS) and the natural defense ability of cells against oxidation.47 As an example, oxidative modifications of proteins by ROS can induce nitrotyrosine expression by endothelial cells and high-level expression of receptors for uremic toxins such as advanced glycation end products (AGE). In the cardiovascular system, AGE accumulation contributes to arterial stiffening due to its binding to collagen and elastin in a disorderly way. Moreover, uremic toxicity leads to an impairment of endothelial nitric oxide (NO) synthesis, which plays a crucial role in vascular protection since NO inhibits the proliferation and migration of VSMC, expression of adhesion molecules, and platelet aggregation.48

The precise relationship of oxidative stress with VC is not yet well established. Yamada et al.49 observed in rats with CKD induced by adeninerich diet a progressive development of arterial medial calcification, which was accompanied by a time-dependent increase in both aortic and systemic oxidative stress. Time-course studies indicated that both oxidative stress and hyperphosphatemia were correlated with arterial medial calcification. Our group showed a significant increase in nitrotyrosine immunostaining in aortas of uremic as compared to non-uremic apoE knock-out mice,50 and Guilgen et al.51 reported same finding in arteries from CKD patients as compared to healthy donors, demonstrating the effective participation of oxidative stress in VC.

INFLAMMATION

Chronic systemic inflammation is a common feature in CKD, caused both by the accumulation of pro-inflammatory compounds related to the markedly decreased glomerular filtration rate, and enhanced production and release of inflammatory cytokines.52

Previous in vivo and in vitro studies showed that inflammation induced intracellular lipid accumulation and foam cell formation by disrupting low-density lipoprotein receptor feedback regulation, exacerbating the progression of atherosclerosis and VC.53,54 Another interesting classical observation was reported by Ketteler et al. in a group of 312 stable patients on hemodialysis the authors observed that serum fetuin-A concentration was lower than in healthy controls and was inversely associated with serum C-reactive protein, and that both were associated with enhanced cardiovascular and all-cause mortality.55

These observations exemplify the essential role of inflammation in vascular calcification and its links with mortality in patients on CKD patients.

PATHOPHYSIOLOGIC LINKS BETWEEN BONE AND VASCULAR CALCIFICATION

In recent years many clinical and experimental studies have demonstrated an association between osteoporosis and VC.56-61 Schulz et al.62 prospectively demonstrated an association between the progression of aortic calcification and decreased bone mineral density. The authors also found that the risk of vertebral and hip fractures was greater in women with aortic calcification. Several studies have extended these findings to patients with CKD in whom coronary and aortic calcification is apparently even more closely associated with reduced bone volume and disturbances in bone remodeling, especially in those with low bone turnover.60,63-65

It has long been known that aging, diabetes mellitus,66 dyslipidemia,67 smoking, and alcohol abuse contribute to both decreased bone mineral density and VC increase.68 However, the association persists after adjusting for some of these factors, suggesting the existence of other mechanisms that have not yet been fully elucidated.

Among the mechanisms proposed some point to various vascular pathologies as the underlying cause. Others have suggested that changes in bone cells could affect the vascular tissue. A third mechanism would include metabolic disorders common to a variety of diseases, either alone or in conjunction with inflammation, such as diabetes mellitus and dyslipidemia which would stimulate both bone resorption and vascular disease.

All organs are endowed with a vascular tree assuring the entry of nutrients and oxygen. Bone tissue is no exception to this rule. In addition, the blood transports bone cell precursors which are involved in bone remodeling and thereby contribute to the integrity of the skeleton. Ischemia caused by intraosseous atherosclerosis can compromise vascularization and favor osteoporosis.69 An association has also been shown between decreased bone mineral density and the presence of peripheral arterial disease.70 A study in women showed that the rate of bone perfusion was markedly reduced in those with osteoporosis as compared to osteopenic women or those with normal bone mass.71

Recently, novel functions of bone tissue have been discovered in addition to already well-known functions such as locomotion, protection of internal organs and participation in the regulation of mineral metabolism. Osteoblasts, through the production of osteocalcin, have been found to participate in the regulation of fat metabolism, energy homeostasis, and insulin secretion and sensitivity, all of which are essential for proper functioning and integrity of the cardiovascular system.72 Osteocalcin regulates insulin gene expression, β-cell proliferation, and adiponectin expression and secretion in adipocytes.73 Both in general population and in patients with CKD serum osteocalcin has been shown to be positively correlated with serum adiponectin.74,75 Moreover, both correlate inversely with arterial stiffness and progression of coronary calcification.74 Leptin, a hormone that regulates adipose tissue mass, is a potent inhibitor of bone formation and also promotes VC.76 In advanced stages of CKD serum leptin levels are generally high. Coen et al.77 demonstrated that uremic patients with high serum leptin levels and low serum PTH levels were prone to develop VC.

The identification of circulating bone cells with potential for VC raised questions as to whether this could be another link.78-80 Both mesenchymal cells from bone marrow and those of hematological lineage can give rise to circulating bone cells with osteogenic potential that could, for example, also home in atherosclerotic lesions and contribute to intima calcification. The ability of these cells to promote VC has as yet only been demonstrated in vitro but not in vivo.81

Recently, two comprehensive reviews about the bone-vascular axis were published by Fadini et al.81 and London.82

DIAGNOSIS

The clinical diagnosis of media calcification is practically impossible by physical examination alone. Its presence is suggested when palpable arteries are detectable when the sphygmomanometer is inflated to a higher level than the true systolic blood pressure. This propaedeutic maneuver is known as the Osler's sign.83

At present, there is no reliable, sufficiently sensitive and specific biomarker for the diagnosis of VC. As described above, in the "Molecular mechanisms of vascular calcification" section, several biomarkers have been shown to be associated with the initiation and/or development of VC. However, it remains unproven that any of these markers reflects calcium phosphate deposition in the arterial wall. It is therefore unclear at present whether they are of any use for clinical practice.

Non-invasive imaging methods are the gold standard. Among the methods available for the detection and quantification of arterial calcification, electron-beam computed tomography (CT) and the more widely accessible multislice CT technique are the most frequently used methods for a precise assessment of the severity of VC and its progression. Results of coronary artery calcification (CAC) are typically reported using the Agatston CAC score, which is based on the product of calcified plaque area and density coefficient. This score has been shown to be predictive of cardiac events. A limitation is the inability to distinguish between intima and media calcification. Moreover, Kristanto et al.84 pointed out that the early onset of calcium deposition remains invisible even with these quantitative techniques and a zero Agatston score does not exclude the presence of incipient coronary calcification.

The Kidney Disease Improving Global Outcomes (KDIGO) 2009 guideline suggests to use cheap lateral abdominal radiography for the semiquantitative assessment of VC and an echocardiogram for the detection of valvular calcification in patients with CKD stages 3 to 5. In clinical routine this technique is a reasonable alternative to costly CT-based imaging methods.85

Kauppila et al.86 developed an abdominal calcification severity scoring system, using lateral lumbar films. The severity of calcified deposits is graded from 0 to 3, separately for the posterior and anterior wall at each vertebral segment, from the first to the fourth lumbar vertebra. Adragao et al.87 developed another scoring system, using pelvis films for iliac and femoral arteries, and hand films for radial and digital arteries. They divided pelvis and hand films into eight sections and graded from 0 to 8 (maximum), depending on the presence or absence of VC (Figure 1). Although these semi-quantitative methods have limited ability to distinguish between the extension and degree of severity of calcification, they are more widely available and less expensive and can be used for cardiovascular risk stratification.


PREVENTION AND TREATMENT

To date, none among a variety of available treatments has been definitively proven to prevent or reverse VC and it is widely admitted that this complication, once established, is irreversible. However, some drugs, including phosphate binders and cinacalcet, have been shown to halt or at least slow the progression of the disease in patients with CKD. Whether this translates into an improvement of clinical outcomes has yet to be demonstrated.85 Therefore, major efforts should be directed at prevention as the main option.

In observational studies hyperphosphatemia, hypercalcemia and extremely high as well as low serum PTH values have been associated with poor outcomes in CKD. As to serum phosphate, even when its serum levels are in the high normal range, it may be associated with VC and mortality, as shown in patients with no known kidney disease and in CKD stage 3 patients.88,89

In CKD stage 4-5 and 5D patients it is recommended at present to bring serum phosphate, calcium and PTH concentrations to target ranges which have been found to be associated with lower rates of VC (Table 2)85 and risks of mortality (Figure 2).90 However, in early stages of CKD these parameters are in the normal range the majority of patients, in contrast to what is observed in later stages.91 Nevertheless, some groups have examined the effects of reducing phosphate overload and/or bringing PTH levels towards the range of normal values in CKD stage 3-4 patients.92


The following paragraphs describe the main options which are presently available for the prevention of VC, with particular focus on phosphate control, as well as the supposedly protective role of some drugs in current use for the treatment of CKDassociated mineral and bone disorder (CKD-MBD). In addition, we discuss therapeutic modalities for new potential targets.

CONTROL OF SERUM PHOSPHATE

Currently, the options available to lower serum phosphate in CKD patients with hyperphosphatemia are (i) limiting dietary phosphate intake (while ensuring adequate protein intake), (ii) increasing the frequency or duration of dialysis in CKD stage 5D, and (iii) using phosphate binders and calcimimetics.

DIET

The KDIGO guideline suggests, in patients with CKD stages 3-5D, to limit dietary phosphate intake in the treatment of hyperphosphatemia alone or in combination with other treatments but this suggestion is mainly based on expert opinion, not on hard evidence.85 In patients with CKD stage 5, phosphate intake should not exceed 1.000 mg per day. Recently, increasing attention has been paid not only to the amount of phosphate in the diet, but also to its quality. Inorganic phosphate in food additives is frequently found in processed food and "fast food". In contrast to organic phosphate, inorganic phosphate is more effectively absorbed and therefore leads to phosphate overload more easily.93 Therefore, the multiprofessional clinical staff who has in charge the treatment of CKD patients should have two goals in mind with respect to recommendations on phosphate intake: avoid inorganic phosphate contained in food additives and limit daily phosphate intake to less than 1.000 mg/day. Note that these goals should be attained without inducing protein malnutrition.

PHOSPHATE REMOVAL BY DIALYSIS

Intradialytic plasma phosphate kinetics obeys to a 2-compartment model, thus differing from urea kinetics. The elimination mode of phosphate appears to resemble more that of typical middle molecules than that of small molecules such as urea.94 Phosphate clearance during hemodialysis is affected by several factors, including blood and dialysate flow rate, dialyzer membrane surface area, ultrafiltration rate, and dialysis session frequency and length.

Increasing dialysis frequency (e.g., short daily dialysis, 6 times a week, for 2.5-3 hr each session) or dialysis length (e.g., nocturnal hemodialysis, 6 times a week, 8 hr each session) are helpful strategies to treat hyperphosphatemia in CKD stage 5D.95,96 Hemodiafiltration can increase phosphate mass removal further.97 In peritoneal dialysis (PD), a cross-sectional comparative study between automated PD and continuous ambulatory PD showed that weekly total phosphate mass removal was similar with both methods.98 Mass transfer could be increased by increasing total daily peritoneal fluid infusion.99 An interesting review on phosphate removal using various hemodialysis and PD treatment modalities was recently published by Kuhlmann.100

ORAL PHOSPHATE BINDERS

Phosphate binder choice should be individualized depending on patients' preference and tolerance. The greatest problem with all phosphate binders is not lack of efficacy, but lack of patient compliance. All currently available phosphate binders, such as calcium or magnesium salts, sevelamer hydrochloride or carbonate, and lanthanum carbonate are effective in lowering serum phosphate.85 Of note, there is insufficient evidence that any specific phosphate binder significantly impacts patient-level outcomes.

Although there is some published evidence suggesting that sevelamer compared with calcium-based phosphate binders attenuates the progression of VC in patients with CKD stages 3-5101 and 5D,102,103 other studies failed to confirm these results.104,105 Sevelamer's effects on VC could be both direct and indirect. This phosphate binder exerts pleiotropic effects, including an attenuation of oxidative stress and inflammation and a decrease in circulating levels of uremic toxins.106,107

Regarding calcium-based phosphate binders, they can lead to calcium overload when taken in excessive amounts.108,109 They should be restricted to no more than a total of 1.500 mg elemental Ca intake per day, especially in presence of VC, hypercalcemia and/or adynamic bone disease,85 the two latter conditions being associated with VC. Magnesium/calcium-based phosphate binders have recently been shown to be an acceptable alternative.110

CALCIMIMETICS

Both experimental and clinical evidence indicates that the calcium-sensing receptor (CaR) is not only expressed in kidney and parathyroid tissue, but also in vascular cells, and that plays a role in VC. In a mouse model of CKD, calcimimetics delayed the progression of aortic calcification and atherosclerosis,111 and in a uremic rat model calcimimetics attenuated media calcification and proliferation of VSMC.112

There is more limited evidence for a positive effect of calcimimetics on VC in the clinical setting. The administration of calcimimetics to dialysis patients with secondary hyperparathyroidism, in addition to lowering serum PTH, calcium and phosphate,113 can also reduce the progression of VC, as shown in the recent ADVANCE study. The authors examined the effect of cinacalcet plus low-dose vitamin D on coronary artery and cardiac valve calcification in 360 prevalent HD patients with secondary hyperparathyroidism, as compared to placebo with optimal standard therapy and flexible doses of vitamin D. In the cinacalcet group there was a 24% increase in Agatston CAC score, as compared to a 31% increase in placebo group (p = 0.073), with corresponding changes in the more recently developed volume CAC score of 22% and 30%, respectively (p = 0.009).114

One of the mechanisms by which a calcimimetic may slow VC progression is its serum PTH, calcium and phosphate lowering action. Other potential mechanisms are a direct stimulation of the CaR expressed in VSMC and an increase in MGP expression in the arterial wall, as shown both in in vivo and in vitro experiments.45,115

VITAMIN D

Both native vitamin D and active vitamin D sterols, also called vitamin D receptor activators (VDRA), may be useful to treat secondary hyperparathyroidism, a condition associated with VC. However, pharmacological doses can result in undesirable effects such as the development of adynamic bone disease,116 hypercalcemia, and/or hyperphosphatemia,117 which all favor the development of VC. Newer VDRAs, such as paricalcitol, have been said to be more selective in suppressing PTH secretion and to be less hypercalcemic and hyperphosphatemic.118

Some experimental studies provided evidence in favor of this claim.119-121 An interesting observation came from the experimental study of Lim et al.40 The authors observed that CKD induces vascular klotho deficiency. They further demonstrated that both calcitriol and paricalcitol significantly upregulated klotho and restored FGF receptor-1 mRNA expression in VSMC of arteries from patients with CKD, which were cultured in procalcific media. They proposed that klotho restoration by vitamin D receptor activation confers VSMC FGF23 responsiveness and unmasks FGF23 calcification inhibitory effects.

Despite these exciting news from experimental models, there is so far no convincing study demonstrating that any vitamin D derivative is less prone than the native parent compounds to induce VC in patients with CKD.

NEW POTENTIAL TREATMENT MODALITIES

PYROPHOSPHATE (PPI)

PPi is a potent calcification inhibitor in vitro and an inhibitor of arterial media calcification in vivo, exerting its effects through direct physicochemical inhibition of hydroxyapatite crystal formation.122,123 Despite its known protective effects against the progression of VC, intravenous administration has been considered to be problematic due to short-half life and complications such as skin necrosis.124 Renewed interest in the calcification inhibitory effects of PPi has been raised by recent experimental studies.125,126 Since hemodialysis patients have low circulating PPi levels,127 an interesting therapeutic option could be to deliver PPi into the peritoneal cavity from where PPi would be slowly transported into the circulation. Riser et al.125 and O'Neill et al.126 have recently shown that daily administration of sodium PPi by peritoneal route was able to prevent the development of aorta calcification in two different animal models of CKD. The next step could be exploratory studies in PD patients.

VITAMIN K

MGP, which is synthesized by VSMC in the arterial media, is a vitamin K-dependent inhibitor of calcium and phosphate precipitation and crystal formation in the vessel wall. In addition, it suppresses the activity of bone morphogenetic proteins 2 and 4.128-130 Vitamin K deficiency affects the activation of MGP by gamma-carboxylation of glutamic acid residues. Undercarboxylated and/or nonphosphorylated MGP loses its inhibitory action on the development and progression of VC and associates with mortality risk in CKD patients.131,132 A recent study showed that the majority of HD patients have a poor vitamin K status and low vitamin K intake compared with healthy subjects.133 Another recent study showed that vitamin K deficiency was associated with fractures and VC in general population.134 In patients with CKD, low vitamin K intake may be related, at least in part, to the dietary regimen generally prescribed, which is restricted in green vegetables and other foods containing large amounts of potassium, but also vitamin K.

To improve vitamin K status and thereby MGP protein activity in CKD patients, vitamin K supplementation may be indicated.135 Thus, Westenfeld et al. observed in stable long-term hemodialysis patients that inactive MGP concentration can be markedly decreased by daily vitamin K supplementation during 6 weeks.136

To date, there are some trials registered on the U.S. National Institutes of Health website (www.clinicaltrials.gov) that are related with vitamin K, CKD and VC. A search with the input keywords "Vitamin K" AND "Chronic kidney disease" resulted in three ongoing trials: "Warfarin and Coronary Calcification Project (WACC)", "Vitamin K to Attenuate Coronary Artery Calcification in Hemodialysis Patients (iPACK HD)", and "Vitamin K2 and Vessel Calcification in Chronic Kidney Disease Patients (CACSK2)". Also, we should mention the European study VITAVASC. We expect to have, in the near future, more information on potentially beneficial effects of vitamin K supplementation on VC risk and outcomes in patients with CKD.

GENERAL CONCLUSION

VC is a complex disease process. It involves not only the local precipitation of calcium and phosphate in the vessel wall, but it also is a regulated cell mediated process, which is under the control of both inhibitory and stimulatory proteins and nonpeptidic factors. This normal, physiological equilibrium is disturbed by CKD, favouring the initiation and progression of VC in parallel with the progressive decline in kidney function.

Based on available experimental and clinical evidence, several drugs currently used for the treatment of CKD-MBD, including sevelamer and calcimimetics, appear to exert at least partially protective effects against the VC process associated with CKD. Drugs such as PPi and vitamin K may represent new avenues for a hopefully more efficient prevention and treatment of VC and its dramatic cardiovascular complications. However, the protective effects of these latter drugs have yet to be demonstrated by future randomized, controlled trials in patients with CKD.

ACKNOWLEDGEMENTS

Financial support. We aknowledge funding by a grant from University of Picardie-Jules Verne. R.B. de Oliveira and H. Okazaki received postdoctoral grants from Picardy Regional Council/University of Picardy Jules Verne, Amiens, France. R.B. de Oliveira and A.E.M. Stinghen received postdoctoral scholarships from the National Council of Technological and Scientific Development (CNPq), Brasília, Brazil.

CONFLICT OF INTEREST STATEMENT

T.B. Drüeke declares having received speaker honoraria, consulting fees and/or research funding from Abbott, Amgen, Fresenius, Genzyme, Kirin, Leo, Mitsubishi, Shire and Theraclion. Z.A. Massy declares having received speaker honoraria, consulting fees and/or research funding from Amgen, Fresenius, Genzyme and Shire. V. Jorgetti declares having received speaker honoraria, consulting fees and/or research funding from Amgen, Abbott and Genzyme.

REFERENCES

Submitted on: 01/30/2013.

Approved on: 03/21/2013.

We acknowledge funding by a grant from University of Picardie-Jules Verne. R.B. de Oliveira and H. Okazaki received postdoctoral grants from Picardy Regional Council/University of Picardy Jules Verne, Amiens, France. R.B. de Oliveira and A.E.M. Stinghen received postdoctoral scholarships from the National Council of Technological and Scientific Development (CNPq), Brasília, Brazil.

  • 1
    Health Science Descriptors [cited 2012 december 07]; 1(1):[1 screen]. Available from: URL: http://decs.bvs.br/
    » link
  • 2. Temmar M, Liabeuf S, Renard C, Czernichow S, Esper NE, Shahapuni I, et al. Pulse wave velocity and vascular calcification at different stages of chronic kidney disease. J Hypertens 2010;28:163-9.
  • 3. Parfitt AM. Soft-tissue calcification in uremia. Arch Intern Med 1969;124:544-56. http://dx.doi.org/10.1001/archinte.124.5.544 http://dx.doi.org/10.1001/archinte.1969.00300210026004 PMid:4899444
  • 4. Steiner I, Laco J. Rokitansky on atherosclerosis. Cesk Patol 2008;44:23-4. PMid:18333331
  • 5. Mönckeberg JG. Ueber die reine Mediaverkalkung der Extremitaetenarterien und ihr Verhalten zur Arteriosklerose. Virchows Arch Pathol Anat 1903;171:141-67. http://dx.doi.org/10.1007/ BF01926946
  • 6. London GM, Guérin AP, Marchais SJ, Métivier F, Pannier B, Adda H. Arterial media calcification in end-stage renal disease: impact on all-cause and cardiovascular mortality. Nephrol Dial Transplant 2003;18:1731-40. http://dx.doi.org/10.1093/ndt/gfg414 PMid:12937218
  • 7. Chiu YW, Adler SG, Budoff MJ, Takasu J, Ashai J, Mehrotra R. Coronary artery calcification and mortality in diabetic patients with proteinuria. Kidney Int 2010;77:1107-14. http://dx.doi.org/10.1038/ki.2010.70 PMid:20237457
  • 8. Scott S, O'Sullivan M, Hafizi S, Shapiro LM, Bennett MR. Human vascular smooth muscle cells from restenosis or in-stent stenosis sites demonstrate enhanced responses to p53: implications for brachytherapy and drug treatment for restenosis Circ Res 2002;90:398-404. http://dx.doi.org/10.1161/hh0402.105900 PMid:11884368
  • 9. Littlewood TD, Bennett MR. Apoptotic cell death in atherosclerosis. Curr Opin Lipidol 2003;14:469-75. http://dx.doi.org/10.1097/00041433-200310000-00007 PMid:14501585
  • 10. Kockx MM. Apoptosis in the atherosclerotic plaque: quantitative and qualitative aspects. Arterioscler Thromb Vasc Biol 1998;18:1519-22. http://dx.doi.org/10.1161/01.ATV.18.10.1519 PMid:9763521
  • 11. Proudfoot D, Skepper JN, Hegyi L, Bennett MR, Shanahan CM, Weissberg PL. Apoptosis regulates human vascular calcification in vitro: evidence for initiation of vascular calcification by apoptotic bodies. Circ Res 2000;87:1055-62. http://dx.doi.org/10.1161/01.RES.87.11.1055 PMid:11090552
  • 12. Shanahan CM, Crouthamel MH, Kapustin A, Giachelli CM. Arterial calcification in chronic kidney disease: key roles for calcium and phosphate. Circ Res 2011;109:697-711. http://dx.doi.org/10.1161/CIRCRESAHA.110.234914 PMid:21885837 PMCid:3249146
  • 13. Shao JS, Cai J, Towler DA. Molecular mechanisms of vascular calcification: lessons learned from the aorta. Arterioscler Thromb Vasc Biol 2006;26:1423-30. http://dx.doi.org/10.1161/01.ATV.0000220441.42041.20 PMid:16601233
  • 14. Bea F, Blessing E, Bennett B, Levitz M, Wallace EP, Rosenfeld ME. Simvastatin promotes atherosclerotic plaque stability in apoE-deficient mice independently of lipid lowering. Arterioscler Thromb Vasc Biol 2002;22:1832-7. http://dx.doi.org/10.1161/01.ATV.0000036081.01231.16 PMid:12426212
  • 15. Speer MY, Yang HY, Brabb T, Leaf E, Look A, Lin WL, et al. Smooth muscle cells give rise to osteochondrogenic precursors and chondrocytes in calcifying arteries. Circ Res 2009;104:733-41. http://dx.doi.org/10.1161/CIRCRESAHA.108.183053 PMid:19197075 PMCid:2716055
  • 16. Tyson KL, Reynolds JL, McNair R, Zhang Q, Weissberg PL, Shanahan CM. Osteo/chondrocytic transcription factors and their target genes exhibit distinct patterns of expression in human arterial calcification. Arterioscler Thromb Vasc Biol 2003;23:489-94. http://dx.doi.org/10.1161/01.ATV.0000059406.92165.31 PMid:12615658
  • 17. Tang Z, Wang A, Yuan F, Yan Z, Liu B, Chu JS, et al. Differentiation of multipotent vascular stem cells contributes to vascular diseases. Nat Commun 2012;3:875. http://dx.doi.org/10.1038/ncomms1867 PMid:22673902 PMCid:3538044
  • 18. Nguyen AT, Gomez D, Bell RD, Campbell JH, Clowes AW, Gabbiani G, et al. Smooth muscle cell plasticity: fact or fiction? Circ Res 2013;112:17-22. http://dx.doi.org/10.1161/CIRCRESAHA.112.281048 PMid:23093573
  • 19. Simionescu A, Philips K, Vyavahare N. Elastin-derived peptides and TGF-beta1 induce osteogenic responses in smooth muscle cells. Biochem Biophys Res Commun 2005;334:524-32. http://dx.doi.org/10.1016/j.bbrc.2005.06.119 PMid:16005428
  • 20. Hosaka N, Mizobuchi M, Ogata H, Kumata C, Kondo F, Koiwa F, et al. Elastin degradation accelerates phosphate-induced mineralization of vascular smooth muscle cells. Calcif Tissue Int 2009;85:523-9. http://dx.doi.org/10.1007/s00223-009-9297-8 PMid:19806384
  • 21. Luo G, Ducy P, McKee MD, Pinero GJ, Loyer E, Behringer RR, et al. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature 1997;386:78-81. http://dx.doi.org/10.1038/386078a0 PMid:9052783
  • 22. Speer MY, McKee MD, Guldberg RE, Liaw L, Yang HY, Tung E, et al. Inactivation of the osteopontin gene enhances vascular calcification of matrix Gla protein-deficient mice: evidence for osteopontin as an inducible inhibitor of vascular calcification in vivo. J Exp Med 2002;196:1047-55. http://dx.doi.org/10.1084/jem.20020911 PMid:12391016 PMCid:2194039
  • 23. Schafer C, Heiss A, Schwarz A, Westenfeld R, Ketteler M, Floege J, et al. The serum protein alpha 2-Heremans-Schmid glycoprotein/fetuin-A is a systemically acting inhibitor of ectopic calcification. J Clin Invest 2003;112:357-66. http://dx.doi.org/10.1172/JCI17202 http://dx.doi.org/10.1172/JCI200317202 PMid:12897203 PMCid:166290
  • 24. Collin-Osdoby P. Regulation of vascular calcification by osteoclast regulatory factors RANKL and osteoprotegerin. Circ Res 2004;95:1046-57. http://dx.doi.org/10.1161/01.RES.0000149165.99974.12 PMid:15564564
  • 25. Hruska KA, Mathew S, Saab G. Bone morphogenetic proteins in vascular calcification. Circ Res 2005;97:105-14. http://dx.doi.org/10.1161/01.RES.00000175571.53833.6c PMid:16037577
  • 26. Balderman JA, Lee HY, Mahoney CE, Handy DE, White K, Annis S, et al. Bone morphogenetic protein-2 decreases microRNA- 30b and microRNA-30c to promote vascular smooth muscle cell calcification. J Am Heart Assoc 2012;1:e003905. PMid:23316327 PMCid:3540659
  • 27. Berndt T, Kumar R. Phosphatonins and the regulation of phosphate homeostasis. Annu Rev Physiol. 2007;69:341-59. http://dx.doi.org/10.1146/annurev.physiol.69.040705.141729 PMid:17002592
  • 28. Son BK, Akishita M, Iijima K, Eto M, Ouchi Y. Mechanism of pi-induced vascular calcification. J Atheroscler Thromb 2008;15:63-8. http://dx.doi.org/10.5551/jat.E545 PMid:18385534
  • 29. Jono S, McKee MD, Murry CE, Shioi A, Nishizawa Y, Mori K, et al. Phosphate regulation of vascular smooth muscle cell calcification. Circ Res 2000;87:E10-7. http://dx.doi.org/10.1161/01.RES.87.7.e10 PMid:11009570
  • 30. Li X, Yang HY, Giachelli CM. Role of the sodium-dependent phosphate cotransporter, Pit-1, in vascular smooth muscle cell calcification. Circ Res 2006;98:905-12. http://dx.doi.org/10.1161/01.RES.0000216409.20863.e7 PMid:16527991
  • 31. Rutsch F, Ruf N, Vaingankar S, Toliat MR, Suk A, Höhne W, et al. Mutations in ENPP1 are associated with 'idiopathic' infantile arterial calcification. Nat Genet 2003;34:379-81. http://dx.doi.org/10.1038/ng1221 PMid:12881724
  • 32. Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T, et al. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest 2004;113:561-8. http://dx.doi.org/10.1172/JCI200419081 http://dx.doi.org/10.1172/JCI19081 PMid:14966565 PMCid:338262
  • 33. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 1997;390:45-51. http://dx.doi.org/10.1038/36285 PMid:9363890
  • 34. Hu MC, Shi M, Zhang J, Qui-ones H, Griffith C, Kuro-o M, et al. Klotho deficiency causes vascular calcification in chronic kidney disease. J Am Soc Nephrol 2011;22:124-36. http://dx.doi.org/10.1681/ASN.2009121311 PMid:21115613 PMCid: 3014041
  • 35. Mirza MA, Hansen T, Johansson L, Ahlström H, Larsson A, Lind L, et al. Relationship between circulating FGF23 and total body atherosclerosis in the community. Nephrol Dial Transplant 2009;24:3125-31. http://dx.doi.org/10.1093/ndt/gfp205 PMid:19429932
  • 36. Jean G, Terrat JC, Vanel T, Hurot JM, Lorriaux C, Mayor B, et al. High levels of serum fibroblast growth factor (FGF)-23 are associated with increased mortality in long haemodialysis patients. Nephrol Dial Transplant 2009;24:2792-6. http://dx.doi.org/10.1093/ndt/gfp191 PMid:19395730
  • 37. Desjardins L, Liabeuf S, Renard C, Lenglet A, Lemke HD, Choukroun G, et al.; European Uremic Toxin (EUTox) Work Group. FGF23 is independently associated with vascular calcification but not bone mineral density in patients at various CKD stages. Osteoporos Int 2012;23:2017-25. http://dx.doi.org/10.1007/s00198-011-1838-0 PMid:22109743
  • 38. Cancela AL, Santos RD, Titan SM, Goldenstein PT, Rochitte CE, Lemos PA, et al. Phosphorus is associated with coronary artery disease in patients with preserved renal function. PLoS One 2012;7:e36883. http://dx.doi.org/10.1371/journal.pone.0036883 PMid:22590632 PMCid:3349637
  • 39. Scialla JJ, Lau WL, Reilly MP, Isakova T, Yang HY, Crouthamel MH, et al. Fibroblast growth factor 23 is not associated with and does not induce arterial calcification. Kidney Int 2013; doi: 10.1038/ki.2013.3. [Epub ahead of print]http://dx.doi.org/10.1038/ki.2013.3
  • 40. Lim K, Lu TS, Molostvov G, Lee C, Lam FT, Zehnder D, et al. Vascular Klotho deficiency potentiates the development of human artery calcification and mediates resistance to fibroblast growth factor 23. Circulation 2012;125:2243-55. http://dx.doi.org/10.1161/CIRCULATIONAHA.111.053405 PMid:22492635
  • 41. Brown EM, MacLeod RJ. Extracellular calcium sensing and extracellular calcium signaling. Physiol Rev 2001;81:239-97. PMid:11152759
  • 42. Smajilovic S, Hansen JL, Christoffersen TE, Lewin E, Sheikh SP, Terwilliger EF, et al. Extracellular calcium sensing in rat aortic vascular smooth muscle cells. Biochem Biophys Res Commun 2006;348:1215-23. http://dx.doi.org/10.1016/j.bbrc.2006.07.192 PMid:16919596
  • 43. Ziegelstein RC, Xiong Y, He C, Hu Q. Expression of a functional extracellular calcium-sensing receptor in human aortic endothelial cells. Biochem Biophys Res Commun 2006;342:153-63. http://dx.doi.org/10.1016/j.bbrc.2006.01.135 PMid:16472767
  • 44. Alam MU, Kirton JP, Wilkinson FL, Towers E, Sinha S, Rouhi M, et al. Calcification is associated with loss of functional calcium-sensing receptor in vascular smooth muscle cells. Cardiovasc Res 2009;81:260-8. http://dx.doi.org/10.1093/cvr/cvn279 PMid:18852253
  • 45. Ivanovski O, Nikolov IG, Joki N, Caudrillier A, Phan O, Mentaverri R, et al. The calcimimetic R-568 retards uremia-enhanced vascular calcification and atherosclerosis in apolipoprotein E deficient (apoE-/-) mice. Atherosclerosis 2009;205:55-62. http://dx.doi.org/10.1016/j.atherosclerosis.2008.10.043 PMid:19118829
  • 46. Koleganova N, Piecha G, Ritz E, Schmitt CP, Gross ML. A calcimimetic (R-568), but not calcitriol, prevents vascular remodeling in uremia. Kidney Int 2009;75:60-71. http://dx.doi.org/10.1038/ki.2008.490 PMid:19092814
  • 47. Small DM, Coombes JS, Bennett N, Johnson DW, Gobe GC. Oxidative stress, anti-oxidant therapies and chronic kidney disease. Nephrology (Carlton) 2012;17:311-21. http://dx.doi.org/10.1111/j.1440-1797.2012.01572.x PMid:22288610
  • 48. Stinghen AE, Pecoits-Filho R. Vascular damage in kidney disease: beyond hypertension. Int J Hypertens 2011;2011:232683.
  • 49. Yamada S, Taniguchi M, Tokumoto M, Toyonaga J, Fujisaki K, Suehiro T, et al. The antioxidant tempol ameliorates arterial medial calcification in uremic rats: important role of oxidative stress in the pathogenesis of vascular calcification in chronic kidney disease. J Bone Miner Res 2012;27:474-85. http://dx.doi.org/10.1002/jbmr.539 PMid:21987400
  • 50. Phan O, Ivanovski O, Nguyen-Khoa T, Mothu N, Angulo J, Westenfeld R, et al. Sevelamer prevents uremia-enhanced atherosclerosis progression in apolipoprotein E-deficient mice. Circulation 2005;112:2875-82. http://dx.doi.org/10.1161/CIRCULATIONAHA105.541854 PMid:16267260
  • 51. Guilgen G, Werneck ML, de Noronha L, Martins AP, Varela AM, Nakao LS, et al. Increased calcification and protein nitration in arteries of chronic kidney disease patients. Blood Purif 2011;32:296-302. http://dx.doi.org/10.1159/000330327 PMid:21876352
  • 52. Kaysen GA. The microinflammatory state in uremia: causes and potential consequences. J Am Soc Nephrol 2001;12:1549-57. PMid:11423586
  • 53. Ruan XZ, Moorhead JF, Tao JL, Ma KL, Wheeler DC, Powis SH, et al. Mechanisms of dysregulation of low-density lipoprotein receptor expression in vascular smooth muscle cells by inflammatory cytokines. Arterioscler Thromb Vasc Biol 2006;26:1150-5. http://dx.doi.org/10.1161/01.ATV.0000217957.93135.c2 PMid:16543490
  • 54. Liu J, Ma KL, Gao M, Wang CX, Ni J, Zhang Y, et al. Inflammation disrupts the LDL receptor pathway and accelerates the progression of vascular calcification in ESRD patients. PLoS One 2012;7:e47217. http://dx.doi.org/10.1371/journal.pone.0047217 PMid:23115640 PMCid:3480367
  • 55. Ketteler M, Bongartz P, Westenfeld R, Wildberger JE, Mahnken AH, Böhm R, et al. Association of low fetuin-A (AHSG) concentrations in serum with cardiovascular mortality in patients on dialysis: a cross-sectional study. Lancet 2003;361:827-33. http://dx.doi.org/10.1016/S0140-6736(03)12710-9
  • 56. Price PA, Roublick AM, Williamson MK. Artery calcification in uremic rats is increased by a low protein diet and prevented by treatment with ibandronate. Kidney Int 2006;70:1577-83. http://dx.doi.org/10.1038/sj.ki.5001841 PMid:16955099
  • 57. Price PA, Faus SA, Williamson MK. Bisphosphonates alendronate and ibandronate inhibit artery calcification at doses comparable to those that inhibit bone resorption. Arterioscler Thromb Vasc Biol 2001;21:817-24. http://dx.doi.org/10.1161/01.ATV.21.5.817 PMid:11348880
  • 58. Hamerman D. Osteoporosis and atherosclerosis: biological linkages and the emergence of dual-purpose therapies. QJM 2005;98:467-84. http://dx.doi.org/10.1093/qjmed/hci077 PMid:15955801
  • 59. Tankó LB, Christiansen C, Cox DA, Geiger MJ, McNabb MA, Cummings SR. Relationship between osteoporosis and cardiovascular disease in postmenopausal women. J Bone Miner Res 2005;20:1912-20. http://dx.doi.org/10.1359/JBMR.050711 PMid:16234963
  • 60. Barreto DV, Barreto FC, Carvalho AB, Cuppari L, Cendoroglo M, Draibe SA, et al. Coronary calcification in hemodialysis patients: the contribution of traditional and uremia-related risk factors. Kidney Int 2005;67:1576-82. http://dx.doi.org/10.1111/j.1523-1755.2005.00239.x PMid:15780114
  • 61. London GM, Marty C, Marchais SJ, Guerin AP, Metivier F, de Vernejoul MC. Arterial calcifications and bone histomorphometry in end-stage renal disease. J Am Soc Nephrol 2004;15:1943-51. http://dx.doi.org/10.1097/01.ASN.0000129337.50739.48 PMid:15213285
  • 62. Schulz E, Arfai K, Liu X, Sayre J, Gilsanz V. Aortic calcification and the risk of osteoporosis and fractures. J Clin Endocrin Metab 2004;89:4246-53. http://dx.doi.org/10.1210/jc.2003-030964 PMid:15356016
  • 63. Adragao T, Herberth J, Monier-Faugere MC, Branscum AJ, Ferreira A, Frazao JM, et al. Low bone volume-a risk factor for coronary calcifications in hemodialysis patients. Clin J Am Soc Nephrol 2009;4:450-5. http://dx.doi.org/10.2215/CJN.01870408 PMid:19158372 PMCid:2637600
  • 64. London GM, Marchais SJ, Guérin AP, Boutouyrie P, Métivier F, de Vernejoul MC. Association of bone activity, calcium load, aortic stiffness, and calcifications in ESRD. J Am Soc Nephrol 2008;19:1827-35. http://dx.doi.org/10.1681/ASN.2007050622 PMid:18480316 PMCid:2518431
  • 65. Toussaint ND, Lau KK, Strauss BJ, Polkinghorne KR, Kerr PG. Associations between vascular calcification, arterial stiffness and bone mineral density in chronic kidney disease. Nephrol Dial Transplant 2008;23:586-93. http://dx.doi.org/10.1093/ndt/gfm660 PMid:17933842
  • 66. Carr JJ, Register TC, Hsu FC, Lohman K, Lenchik L, Bowden DW, et al. Calcified atherosclerotic plaque and bone mineral density in type 2 diabetes: the diabetes heart study. Bone 2008;42:43-52. http://dx.doi.org/10.1016/j.bone.2007.08.023 PMid:17964237 PMCid:2239236
  • 67. Demer L, Tintut Y. The roles of lipid oxidation products and receptor activator of nuclear factor-κB signaling in atherosclerotic calcification. Circ Res 2011;108:1482-93. http://dx.doi.org/10.1161/CIRCRESAHA.110.234245 PMid:21659652 PMCid:3128471
  • 68. Kulak CA, Borba VC, Jorgetti V, Dos Reis LM, Liu XS, Kimmel DB, et al. Skeletal microstructural abnormalities in postmenopausal women with chronic obstructive pulmonary disease. J Bone Miner Res 2010;25:1931-40. http://dx.doi.org/10.1002/jbmr.88 PMid:20564248
  • 69. Bridgeman G, Brookes M. Blood supply to the human femoral diaphysis in youth and senescence. J Anat 1996;188:611-21. PMid:8763478 PMCid:1167489
  • 70. Laroche M. Intraosseous circulation from physiology to disease. Joint Bone Spine 2002;69:262-9. http://dx.doi.org/10.1016/S1297-319X(02)00391-3
  • 71. Griffith JF, Yeung DK, Tsang PH, Choi KC, Kwok TC, Ahuja AT, et al. Compromised bone marrow perfusion in osteoporosis. J Bone Miner Res 2008;23:1068-75. http://dx.doi.org/10.1359/jbmr.080233 PMid:18302498
  • 72. Lee NK, Sowa H, Hinoi E, Ferron M, Ahn JD, Confavreux C, et al. Endocrine regulation of energy metabolism by the skeleton. Cell 2007;130:456-69. http://dx.doi.org/10.1016/j.cell.2007.05.047 PMid:17693256 PMCid:2013746
  • 73. Ferron M, Hinoi E, Karsenty G, Ducy P. Osteocalcin differentially regulates beta cell and adipocyte gene expression and affects the development of metabolic diseases in wild-type mice. Proc Natl Acad Sci U S A 2008;105:5266-70. http://dx.doi.org/10.1073/pnas.0711119105 PMid:18362359 PMCid: 2278202
  • 74 Kanazawa I, Yamaguchi T, Yamamoto M, Yamauchi M, Kurioka S, Yano S, et al. Serum osteocalcin level is associated with glucose metabolism and atherosclerosis parameters in type 2 diabetes mellitus. J Clin Endocrinol Metab 2009;94:45-9. http://dx.doi.org/10.1210/jc.2008-1455 PMid:18984661
  • 75. Bacchetta J, Boutroy S, Guebre-Egziabher F, Juillard L, Drai J, Pelletier S, et al. The relationship between adipokines, osteocalcin and bone quality in chronic kidney disease. Nephrol Dial Transplant 2009;24:3120-5. http://dx.doi.org/10.1093/ndt/gfp262 PMid:19515806
  • 76. Elefteriou F, Takeda S, Ebihara K, Magre J, Patano N, Kim CA, et al. Serum leptin level is a regulator of bone mass. Proc Natl Acad Sci U S A 2004;101:3258-63. http://dx.doi.org/10.1073/pnas.0308744101 PMid:14978271 PMCid:365777
  • 77. Coen G, Ballanti P, Fischer MS, Balducci A, Calabria S, Colamarco L, et al. Serum leptin in dialysis renal osteodystrophy. Am J Kidney Dis 2003;42:1036-42. http://dx.doi.org/10.1016/j.ajkd.2003.07.005 PMid:14582047
  • 78. Eghbali-Fatourechi GZ, Lamsam J, Fraser D, Nagel D, Riggs BL, Khosla S. Circulating osteoblast-lineage cells in humans. N Engl J Med 2005;352:1959-66. http://dx.doi.org/10.1056/NEJMoa044264 PMid:15888696
  • 79. Pirro M, Leli C, Fabbriciani G, Manfredelli MR, Callarelli L, Bagaglia F, et al. Association between circulating osteoprogenitor cell numbers and bone mineral density in postmenopausal osteoporosis. Osteoporos Int 2010;21:297-306. http://dx.doi.org/10.1007/s00198-009-0968-0 PMid:19484167
  • 80. Pal SN, Rush C, Parr A, Van Campenhout A, Golledge J. Osteocalcin positive mononuclear cells are associated with the severity of aortic calcification. Atherosclerosis 2010;210:88-93. http://dx.doi.org/10.1016/j.atherosclerosis.2009.11.001 PMid:20004897 PMCid:2862100
  • 81. Fadini GP, Rattazzi M, Matsumoto T, Asahara T, Khosla S. Emerging role of circulating calcifying cells in the bone-vascular axis. Circulation 2012;125:2772-81. http://dx.doi.org/10.1161/CIRCULATIONAHA.112.090860 PMid:22665885
  • 82. London GM. Bone-vascular cross-talk. J Nephrol 2012;25:619-25.
  • 83. Cunha UGV, Valle EA, Melo RA. Peculiaridades do exame físico do idoso. Rev Med Minas Gerais 2011;21:181-5.
  • 84. Kristanto W, van Ooijen PM, Groen JM, Vliegenthart R, Oudkerk M. Small calcified coronary atherosclerotic plaque simulation model: minimal size and attenuation detectable by 64-MDCT and MicroCT. Int J Cardiovasc Imaging 2012;28:843-53. http://dx.doi.org/10.1007/s10554-011-9869-3 PMid:21509430 PMCid:3360866
  • 85. Kidney Disease: Improving Global Outcomes (KDIGO) CKD- -MBD Work Group. KDIGO clinical practice guideline for the diagnosis, evaluation, prevention, and treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD). Kidney Int Suppl 2009;(113)S1:130.
  • 86. Kauppila LI, Polak JF, Cupples LA, Hannan MT, Kiel DP, Wilson PW. New indices to classify location, severity and progression of calcific lesions in the abdominal aorta: a 25-year follow-up study. Atherosclerosis 1997;132:245-50. http://dx.doi.org/10.1016/S0021-9150(97)00106-8
  • 87. Adragao T, Pires A, Lucas C, Birne R, Magalhaes L, Gonçalves M, et al. A simple vascular calcification score predicts cardiovascular risk in haemodialysis patients. Nephrol Dial Transplant 2004;19:1480-8. http://dx.doi.org/10.1093/ndt/gfh217 PMid:15034154
  • 88. Tomiyama C, Higa A, Dalboni MA, Cendoroglo M, Draibe SA, Cuppari L, et al. The impact of traditional and non-traditional risk factors on coronary calcification in pre-dialysis patients. Nephrol Dial Transplant 2006;21:2464-71. http://dx.doi.org/10.1093/ndt/gfl291 PMid:16735378
  • 89. Kestenbaum B, Sampson JN, Rudser KD, Patterson DJ, Seliger SL, Young B, et al. Serum phosphate levels and mortality risk among people with chronic kidney disease. J Am Soc Nephrol 2005;16:520-8. http://dx.doi.org/10.1681/ASN.2004070602 PMid:15615819
  • 90. Floege J, Kim J, Ireland E, Chazot C, Drueke T, de Francisco A, et al.; ARO Investigators. Serum iPTH, calcium and phosphate, and the risk of mortality in a European haemodialysis population. Nephrol Dial Transplant 2011;26:1948-55. http://dx.doi.org/10.1093/ndt/gfq219 PMid:20466670 PMCid:3107766
  • 91. Levin A, Bakris GL, Molitch M, Smulders M, Tian J, Williams LA, et al. Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney disease. Kidney Int 2007;71:31-8. http://dx.doi.org/10.1038/sj.ki.5002009 PMid:17091124
  • 92. Oliveira RB, Cancela AL, Graciolli FG, Dos Reis LM, Draibe SA, Cuppari L, et al. Early control of PTH and FGF23 in normophosphatemic CKD patients: a new target in CKD-MBD therapy? Clin J Am Soc Nephrol 2010;5:286-91. http://dx.doi.org/10.2215/CJN.05420709 PMid:19965540 PMCid:2827593
  • 93. Ritz E, Hahn K, Ketteler M, Kuhlmann MK, Mann J. Phosphate additives in food--a health risk. Dtsch Arztebl Int 2012;109:49-55. PMid:22334826 PMCid:3278747
  • 94. Bammens B, Evenepoel P, Verbeke K, Vanrenterghem Y. Removal of middle molecules and protein-bound solutes by peritoneal dialysis and relation with uremic symptoms. Kidney Int 2003;64:2238-43. http://dx.doi.org/10.1046/j.1523-1755.2003.00310.x PMid:14633148
  • 95. Ayus JC, Mizani MR, Achinger SG, Thadhani R, Go AS, Lee S. Effects of short daily versus conventional hemodialysis on left ventricular hypertrophy and inflammatory markers: a prospective, controlled study. J Am Soc Nephrol 2005;16:2778-88. http://dx.doi.org/10.1681/ASN.2005040392 PMid:16033855
  • 96. Pierratos A, Ouwendyk M, Francoeur R, Vas S, Raj DS, Ecclestone AM, et al. Nocturnal hemodialysis: three-year experience. J Am Soc Nephrol 1998;9:859-68. PMid:9596084
  • 97. Feliciani A, Riva MA, Zerbi S, Ruggiero P, Plati AR, Cozzi G, et al. New strategies in haemodiafiltration (HDF): prospective comparative analysis between on-line mixed HDF and mid-dilution HDF. Nephrol Dial Transplant 2007;22:1672-9. http://dx.doi.org/10.1093/ndt/gfm023 PMid:17347283
  • 98. Evenepoel P, Bammens B, Verbeke K, Vanrenterghem Y. Superior dialytic clearance of beta(2)-microglobulin and p-cresol by high-flux hemodialysis as compared to peritoneal dialysis. Kidney Int 2006;70:794-9. http://dx.doi.org/10.1038/sj.ki.5001640 PMid:16820785
  • 99. Messa P, Gropuzzo M, Cleva M, Boscutti G, Mioni G, Cruciatti A, et al. Behaviour of phosphate removal with different dialysis schedules. Nephrol Dial Transplant 1998;13:43-8. http://dx.doi.org/10.1093/ndt/13.suppl_6.43 PMid:9719204
  • 100. Kuhlmann MK. Phosphate elimination in modalities of hemodialysis and peritoneal dialysis. Blood Purif 2010;29:137-44. http://dx.doi.org/10.1159/000245640 PMid:20093819
  • 101. Chertow GM, Burke SK, Raggi P.; Treat to Goal Working Group. Sevelamer attenuates the progression of coronary and aortic calcification in hemodialysis patients. Kidney Int 2002;62:245-52. http://dx.doi.org/10.1046/j.1523-1755.2002.00434.x PMid:12081584
  • 102. Block GA, Spiegel DM, Ehrlich J, Mehta R, Lindbergh J, Dreisbach A, et al. Effects of sevelamer and calcium on coronary artery calcification in patients new to hemodialysis. Kidney Int 2005;68:1815-24. http://dx.doi.org/10.1111/j.1523-1755.2005.00600.x PMid:16164659
  • 103. Russo D, Miranda I, Ruocco C, Battaglia Y, Buonanno E, Manzi S, et al. The progression of coronary artery calcification in predialysis patients on calcium carbonate or sevelamer. Kidney Int 2007;72:1255-61. http://dx.doi.org/10.1038/sj.ki.5002518 PMid:17805238
  • 104. Qunibi W, Moustafa M, Muenz LR, He DY, Kessler PD, Diaz-Buxo JA, et al. A 1-year randomized trial of calcium acetate versus sevelamer on progression of coronary artery calcification in hemodialysis patients with comparable lipid control: the Calcium Acetate Renagel Evaluation-2 (CARE-2) study. Am J Kidney Dis 2008;51:952-65. http://dx.doi.org/10.1053/j.ajkd.2008.02.298 PMid:18423809
  • 105. Barreto DV, Barreto Fde C, de Carvalho AB, Cuppari L, Draibe SA, Dalboni MA, et al. Phosphate binder impact on bone remodeling and coronary calcification--results from the BRiC study. Nephron Clin Pract 2008;110c:273-83. http://dx.doi.org/10.1159/000170783 PMid:19001830
  • 106. Vlassara H, Uribarri J, Cai W, Goodman S, Pyzik R, Post J, et al. Effects of sevelamer on HbA1c, inflammation, and advanced glycation end products in diabetic kidney disease. Clin J Am Soc Nephrol 2012;7:934-42. http://dx.doi.org/10.2215/CJN.12891211 PMid:22461535
  • 107. Nikolov IG, Joki N, Maizel J, Lacour B, Drüeke TB, Massy ZA. Pleiotropic effects of the non-calcium phosphate binder sevelamer. Kidney Int Suppl 2006;(105):S16-23. http://dx.doi.org/10.1038/sj.ki.5001994 PMid:17136111
  • 108. Spiegel DM, Brady K. Calcium balance in normal individuals and in patients with chronic kidney disease on low- and high-calcium diets. Kidney Int 2012;81:1116-22. http://dx.doi.org/10.1038/ki.2011.490 PMid:22297674 PMCid:3352985
  • 109. Hill KM, Martin BR, Wastney ME, McCabe GP, Moe SM, Weaver CM, et al. Oral calcium carbonate affects calcium but not phosphorus balance in stage 3-4 chronic kidney disease. Kidney Int 2012; doi: 10.1038/ki.2012.403. [Epub ahead of print]http://dx.doi.org/10.1038/ki.2012.403
  • 110. Drüeke TB, Massy ZA. Phosphate binders in CKD: bad news or good news? J Am Soc Nephrol. 2012;23:1277-80. http://dx.doi.org/10.1681/ASN.2012060569 PMid:22797178
  • 111. Joki N, Nikolov IG, Caudrillier A, Mentaverri R, Massy ZA, Drüeke TB. Effects of calcimimetic on vascular calcification and atherosclerosis in uremic mice. Bone 2009;45:S30-4. http://dx.doi.org/10.1016/j.bone.2009.03.653 PMid:19303957
  • 112. Koleganova N, Piecha G, Ritz E, Schmitt CP, Gross ML. A calcimimetic (R-568), but not calcitriol, prevents vascular remodeling in uremia. Kidney Int 2009;75:60-71. http://dx.doi.org/10.1038/ki.2008.490 PMid:19092814
  • 113. Torres PA, De Broe M. Calcium-sensing receptor, calcimimetics, and cardiovascular calcifications in chronic kidney disease. Kidney Int 2012;82:19-25. http://dx.doi.org/10.1038/ki.2012.69 PMid:22437409
  • 114. Raggi P, Chertow GM, Torres PU, Csiky B, Naso A, Nossuli K, et al.; ADVANCE Study Group. The ADVANCE study: a randomized study to evaluate the effects of cinacalcet plus low-dose vitamin D on vascular calcification in patients on hemodialysis. Nephrol Dial Transplant 2011;26:1327-39. http://dx.doi.org/10.1093/ndt/gfq725 PMid:21148030
  • 115. Mendoza FJ, Martinez-Moreno J, Almaden Y, Rodriguez-Ortiz ME, Lopez I, Estepa JC, et al. Effect of calcium and the calcimimetic AMG 641 on matrix-Gla protein in vascular smooth muscle cells. Calcif Tissue Int 2011;88:169-78. http://dx.doi.org/10.1007/s00223-010-9442-4 PMid:21161195
  • 116. Goodman WG, Ramirez JA, Belin TR, Chon Y, Gales B, Segre GV, et al. Development of adynamic bone in patients with secondary hyperparathyroidism after intermittent calcitriol therapy. Kidney Int 1994;46:1160-6. http://dx.doi.org/10.1038/ki.1994.380 PMid:7861712
  • 117. Quarles LD, Yohay DA, Carroll BA, Spritzer CE, Minda SA, Bartholomay D, et al. Prospective trial of pulse oral versus intravenous calcitriol treatment of hyperparathyroidism in ESRD. Kidney Int 1994;45:1710-21. http://dx.doi.org/10.1038/ki.1994.223 PMid:7933819
  • 118. Drüeke TB. Which vitamin D derivative to prescribe for renal patients. Curr Opin Nephrol Hypertens 2005;14:343-9. http://dx.doi.org/10.1097/01.mnh.0000172720.34229.39 PMid:15931002
  • 119. Cardús A, Panizo S, Parisi E, Fernandez E, Valdivielso JM. Differential effects of vitamin D analogs on vascular calcification. J Bone Miner Res 2007;22:860-6. http://dx.doi.org/10.1359/jbmr.070305 PMid:17352647
  • 120. Mizobuchi M, Finch JL, Martin DR, Slatopolsky E. Differential effects of vitamin D receptor activators on vascular calcification in uremic rats. Kidney Int 2007;72:709-15. http://dx.doi.org/10.1038/sj.ki.5002406 PMid:17597697
  • 121. Lau WL, Leaf EM, Hu MC, Takeno MM, Kuro-o M, Moe OW, et al. Vitamin D receptor agonists increase klotho and osteopontin while decreasing aortic calcification in mice with chronic kidney disease fed a high phosphate diet. Kidney Int 2012;82:1261-70. http://dx.doi.org/10.1038/ki.2012.322 PMid:22932118
  • 122. Lomashvili KA, Cobbs S, Hennigar RA, Hardcastle KI, O'Neill WC. Phosphate-induced vascular calcification: role of pyrophosphate and osteopontin. J Am Soc Nephrol 2004;15:1392-401. http://dx.doi.org/10.1097/01.ASN.0000128955.83129.9C PMid:15153550
  • 123. Schibler D, Russell RG, Fleisch H. Inhibition by pyrophosphate and polyphosphate of aortic calcification induced by vitamin D3 in rats. Clin Sci 1968;35:363-72. PMid:4305530
  • 124. Jung A, Russel RG, Bisaz S, Morgan DB, Fleisch H. Fate of intravenously injected pyrophosphate-32P in dogs. Am J Physiol 1970;218:1757-64. PMid:4317203
  • 125. Riser BL, Barreto FC, Rezg R, Valaitis PW, Cook CS, White JA, et al. Daily peritoneal administration of sodium pyrophosphate in a dialysis solution prevents the development of vascular calcification in a mouse model of uraemia. Nephrol Dial Transplant 2011;26:3349-57. http://dx.doi.org/10.1093/ndt/gfr039 PMid:21398365
  • 126. O'Neill WC, Lomashvili KA, Malluche HH, Faugere MC, Riser BL. Treatment with pyrophosphate inhibits uremic vascular calcification. Kidney Int 2011;79:512-7. http://dx.doi.org/10.1038/ki.2010.461 PMid:21124302 PMCid:3183997
  • 127. Lomashvili KA, Khawandi W, O'Neill WC. Reduced plasma pyrophosphate levels in hemodialysis patients. J Am Soc Nephrol 2005;16:2495-500. http://dx.doi.org/10.1681/ASN.2004080694 PMid:15958726
  • 128. Luo G, Ducy P, McKee MD, Pinero GJ, Loyer E, Behringer RR, et al. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature 1997;386:78-81. http://dx.doi.org/10.1038/386078a0 PMid:9052783
  • 129. Wajih N, Borras T, Xue W, Hutson SM, Wallin R. Processing and transport of matrix gamma-carboxyglutamic acid protein and bone morphogenetic protein-2 in cultured human vascular smooth muscle cells: evidence for an uptake mechanism for serum fetuin. J Biol Chem 2004;279:43052-60. http://dx.doi.org/10.1074/jbc.M407180200 PMid:15280384
  • 130. Murshed M, Schinke T, McKee MD, Karsenty G. Extracellular matrix mineralization is regulated locally; different roles of two gla-containing proteins. J Cell Biol 2004;165:625-30. http://dx.doi.org/10.1083/jcb.200402046 PMid:15184399 PMCid:2172384
  • 131. Schurgers LJ, Barreto DV, Barreto FC, Liabeuf S, Renard C, Magdeleyns EJ, et al. The circulating inactive form of matrix gla protein is a surrogate marker for vascular calcification in chronic kidney disease: a preliminary report. Clin J Am Soc Nephrol 2010;5:568-75. http://dx.doi.org/10.2215/CJN.07081009 PMid:20133489 PMCid:2849687
  • 132. Schlieper G, Westenfeld R, Krüger T, Cranenburg EC, Magdeleyns EJ, Brandenburg VM, et al. Circulating nonphosphorylated carboxylated matrix gla protein predicts survival in ESRD. J Am Soc Nephrol 2011;22:387-95. http://dx.doi.org/10.1681/ASN.2010040339 PMid:21289218 PMCid: 3029911
  • 133. Cranenburg EC, Schurgers LJ, Uiterwijk HH, Beulens JW, Dalmeijer GW, Westerhuis R, et al. Vitamin K intake and status are low in hemodialysis patients. Kidney Int 2012;82:605-10. http://dx.doi.org/10.1038/ki.2012.191 PMid:22648294
  • 134. Fusaro M, Noale M, Viola V, Galli F, Tripepi G, Vajente N, Plebani M, et al.; VItamin K Italian (VIKI) Dialysis Study Investigators. Vitamin K, vertebral fractures, vascular calcifications, and mortality: VItamin K Italian (VIKI) dialysis study. J Bone Miner Res 2012;27:2271-8. http://dx.doi.org/10.1002/jbmr.1677 PMid:22692665
  • 135. Krueger T, Westenfeld R, Ketteler M, Schurgers LJ, Floege J. Vitamin K deficiency in CKD patients: a modifiable risk factor for vascular calcification? Kidney Int 2009;76:18-22. http://dx.doi.org/10.1038/ki.2009.126 PMid:19387474
  • 136. Westenfeld R, Krueger T, Schlieper G, Cranenburg EC, Magdeleyns EJ, Heidenreich S, et al. Effect of vitamin K2 supplementation on functional vitamin K deficiency in hemodialysis patients: a randomized trial. Am J Kidney Dis 2012;59:186-95. http://dx.doi.org/10.1053/j.ajkd.2011.10.041 PMid:22169620
  • Correspondence to:

    Vanda Jorgetti
    Division of Nephrology, University of São Paulo, São Paulo, Brazil
    Av. Dr. Arnaldo, nº 455, 3º andar, sala 334
    São Paulo, SP, Brazil. CEP: 01246-903
    Tel: +55 (11) 3061-8351
  • Publication Dates

    • Publication in this collection
      24 June 2013
    • Date of issue
      June 2013

    History

    • Received
      30 Jan 2013
    • Accepted
      21 Mar 2013
    Sociedade Brasileira de Nefrologia Rua Machado Bittencourt, 205 - 5ºandar - conj. 53 - Vila Clementino - CEP:04044-000 - São Paulo SP, Telefones: (11) 5579-1242/5579-6937, Fax (11) 5573-6000 - São Paulo - SP - Brazil
    E-mail: bjnephrology@gmail.com