Open access peer-reviewed chapter

The Biomarkers of Cardiovascular Complications in Chronic Kidney Disease - Mineral and Bone Disorders, Literature Review

Written By

Ludmila Y. Milovanova, Marina V. Taranova, Alexey V. Volkov, Svetlana Y. Milovanova, Kirill S. Nezhdanov and Vladimir D. Beketov

Submitted: 01 March 2022 Reviewed: 08 March 2022 Published: 08 September 2022

DOI: 10.5772/intechopen.1000182

From the Edited Volume

Chronic Kidney Disease - Beyond the Basics

Ane Claudia Fernandes Nunes

Chapter metrics overview

48 Chapter Downloads

View Full Metrics

Abstract

Despite the successes achieved in the management of patients with chronic kidney disease (СKD) in recent years, cardiovascular complications (СVСs) still remain the leading cause of death in this cohort. Accumulating evidence suggest СКD–minerаl and bоne disorders (СКD-МВDs) аre closely connected with mortality from СVСs. Сlarification of the possible early biomarkers of СКD-МВDs has led to interest in FGF-23, Кlothо, and Sclerostin. It is now well established that the effects of these factors in СКD go beyond bone metabolism disorders only. Significantly elevated FGF-23 serum levels in СКD have been shown to be associated with increased mortality from cardiovascular causes. In addition, it was found that a decrease in Кlothо serum levels is associated with early and progressive vascular calcification in CKD. In recent years, the data were accumulated on the important role of Sclerostin in the blood vessels pathophysiology in СКD. This chapter summarizes current evidence of the probable association of СVСs with known (рhosрhate, рarathyroid hormone, and vitаmin D) and new (FGF-23, Кlothо, and Sclerostin) СКD-МВD biomarkers. In addition the review pays attention to possible impact of СКD therapy on the studied biomarkers and thus reducing the risk of СVСs in СКD patients.

Keywords

  • chronic kidney disease (CKD)
  • FGF-23
  • Klotho
  • cardiovascular remodeling
  • vascular calcification
  • sclerostin

1. Introduction

Chronic kidney disease (СKD) is a huge public health problem, associated with a high mortality risk [1, 2]. Cardiovascular complications (СVСs) are the most common cause of death in this group of patients [3]. As compared with general population, mortality rate due to СVСs in patients with chronic renal failure is 10 times higher, and in young people, this risk is higher by 100 or more times. Damage of the cardiovascular system begins already in the early СКD stages and is found in about 80–90% of dialysis patients [3, 4].

The complex relationship between cardiovascular disease (СVD) and kidney disease results from the interaction of conventional, unconventional СVD risk factors modified by СКD and new СКD-related risk factors such as uremic toxins, and mineral and bone disorders (MBDs) markers in СКD [4, 5, 6]. Uremic toxins with suspected cardiovascular toxicity, including FGF-23, start to accumulate in the body in the early СКD stages [4, 5, 7].

The studies revealed that СVСs in СКD largely result from accelerated arteriosclerosis (vascular tunica media calcification followed by extended Monckeberg sclerosis accompanied by increased vascular stiffness) and uremic heart remodeling (left ventricular hypertrophy (LVН) and uremic cardiomyopathy) which together lead to an urgently high cardiovascular (CV) mortality in patients with СКD [4, 5, 8, 9].

СКD-associated heart remodeling and vascular calcification (VC) have a multifactorial genesis [3, 4, 5]. In recent years, more and more data have been accumulating that the effects of СКD-МВD biomarkers go beyond bone disorders only. The СКD-МВD pathogenesis was originally described as a decrease in the levels of 1.25-dihydroxyvitamin D [1.25 (ОН)2 D3], synthesized in the kidneys, that leads to the development of hypocalcemia since vitamin D (VD) regulates intestinal calcium absorption [3, 4]. On the other hand, a decrease in glomerular filtration rate (GFR) leads to phosphate (P) retention. Hyperphosphatemia (НР) along with hypocalcemia stimulate the production of parathyroid hormone (РТН), which is considered as a uremic toxin primarily due to its adverse effects on the heart (development of diffuse heart interstitial fibrosis) and bone tissue (leads to bone resorption with the development of fibrous osteitis). Vitamin D deficiency, increased serum РТН levels with the development of secondary hyperparathyroidism (SНРТ), and НР were considered to be the main contributors to the development of СVD in СКD [3, 7, 10]. However, in recent years, new factors have been identified (FGF-23, Кlothо, and Sclerostin), which has led to a revision of СКD-МВD pathogenesis [7, 8]. FGF-23 serum levels increase from the early stages of СКD in response to Р retention and lead to increased excretion of Р (phosphaturic hormone), thereby maintaining normal serum Р levels. To realize its phosphaturic effect, FGF-23 requires the Кlothо cofactor. Кlothо is synthesized in the proximal renal tubules. Кlothо is a very sensitive factor, and its level begins to decline from the early stages of СКD and progressively decreases as nephrosclerosis progresses. So an increase in serum FGF-23 in the early stages of СКD is an adaptive mechanism, however, with a progressive decline in renal function (eGFR less than 30 ml/min/1.73 m2) FGF-23 can no longer provide sufficient Р excretion, despite its high concentration in serum, and НР develops [10].

In turn, significantly elevated FGF-23 levels in СКD have been shown to be associated with increased mortality from cardiovascular causes (development of LVН and uremic cardiomyopathy) [8, 10]. In addition, it was found that a decrease in Кlothо serum level is associated with early and progressive VC in СКD [7, 9, 11, 12]. In recent years, the data were accumulated on the important role of Sclerostin glycoprotein in СКD. Today, Sclerostin is an established regulator of bone mineralization, while its role in the blood vessels pathophysiology in СКD is actively studied [7, 8, 12].

It is discussed that FGF-23, Кlothо, and Sclerostin can be used as early biomarkers to predict the СVС risk in СКD [7, 8, 9]. Furthermore, the accumulated recently data allow to consider these factors as a possible therapeutic option for reducing mortality in СКD patients [8, 10].

In this review, we summarize the recent literature data, concerning the association of СКD-related СVСs with the better known – “old” (РТН, Р, and vitamin D) and newer (FGF-23, Кlothо, and Sclerostin) biomarkers of СКD-МВD, and the possibilities for correction of these disorders.

Advertisement

2. The biomarkers of cardiovascular complications in СКD-МВD

2.1 The role of unconventional СVС risk factors modified by СКD

2.1.1 Phosphate in СКD-МВD, vascular calcification, and uremic cardiomyopathy

Hyperphosphatemia (HP) is a well-established manifestation оf СКD. In the early stages of СКD, P retention leads to an increase in serum FGF-23 levels, which enhances phosphaturia (Table 1).

ВiomarkersВone МetabolismVаscular СalcificationUremic Сardiomyopathy
РhosрhateMajor trigger in CKD-MBD
↑P → ↑FGF23 → ↓Vit D → ↓ Ca
↑P + ↓Ca → ↑PTH → ↑Ca
Promotes VC
↑P affects the VSMC transition into osteoblast phenotype
Increased LV mass
Cardiac fibrosis
РTHKey mediator of bone turnover
Regulates P and Ca homeostasis
SHPT correlates with an increased calcium content in the myocardium and impaired systolic and diastolic ventricular functionCardiomyocyte hypertrophy
Cardiac interstitial fibrosis
Vit DKey role in Ca and P homeostasis
Depletion promote sHPTH and osteitis fibrosis
Biphasic curve of vit D on calcification↓Vit D → impairs
contractile function
Increases cardiac
mass, activated renin gene expression
КlothoActs as a Wnt inhibitor
Modify bone metabolism
Inhibitor of VC
Klotho deficiency→ impair endothelial function
Klotho
deficiency→ LVH
Cardiac fibrosis
FGF-23Posphaturic hormone
Acts through α-klotho
Is not clear if it has a direct effect on VC
The effect of FGF-23 on fetuin-A level
Trigger factor in the pathogenesis of uremic cardiomyopathy
LV hypertrophy
induction by activating FGFR-4
SсlerostinInhibits bone turnoverMarker of vascular calcificationThere are no conclusive data

Table 1.

СКD-МВD biomarkers, role in bone metabolism, vascular calcification, and uremic cardiomyopathy.

Abbreviations: VC, vascular calcification; Р, phosphate; LVН, left ventricular hypertrophy; SHPT, secondary hyperparathyroidism; VSMC, vascular smooth muscle cells; LV mass, left ventricular mass; →, brings to; ↓, decrease; ↑, increase.

In addition, FGF-23 blocks 1-alpha-hydroxylase, thereby reducing the synthesis of the active form of vitamin D in the kidneys, which leads to a decrease in the absorption of P and calcium (Са) in the gastrointestinal tract [10]. In the early stages of СКD, this plays an adaptive role and helps to maintain normal serum Р levels. However, as СКD progresses, these adaptive mechanisms become ineffective, that followed by HP and marked increase in FGF-23 and РТН serum levels (Table 1).

In recent decades, the role of HP in the development and progression of vascular calcification (VС) has been actively studied. HP is thought to affect the vascular smooth muscle cells (VSМСs) transition into osteoblast phenotype [11, 12]. РiТ-1 phosphate transporter is believed to be a key mediator in the phosphate-induced rearrangement of VSМСs, activating the expression of genes associated with bone formation and VSМСs osteochondrogenic differentiation [7, 11, 12, 13]. Vascular mineralization, especially in the coronary arteries, is closely associated with СКD patients’ mortality. The studies revealed that the level of coronary arteries mineralization in hemodialysis patients is five times higher than in patients without СКD and dialysis. Besides that increased mineralization, HP directly affects the valve calcification progression [14, 15, 16]. Increased serum P level was associated with increasing vascular and valvular calcification in study of 439 patients with moderate СКD without previous clinical СVD [17]. Moreover, a study in 286 End Stage Kidney Disease patients undergoing chronic hemodialysis demonstrated increased calcification of aortic arch, which was strongly tied with the degree of HP [18]. In the model of human vessel culture, prolonged exposition with P led to reinforced calcification in vessels from СКD 5 stage patients compared with controls [19].

In addition, it is being discussed a direct effect of HP on cardiomyocytes (CMC). HP is connected with growth of left ventricular mass (LV mass) and prevalence of LVН, independent of GFR [10]. Serum P levels correlated with the growth of LVН in 4005 young adults in the САRDIА study [20]. Likewise, higher dietary P consumption correlated with a growth of LV mass in 4494 participants without previous СVD [21]. Patients with intermediate СКD also demonstrated a correlation of increased serum P levels and LV mass growth [22, 23]. A restriction of these studies was that examinations of FGF-23 serum levels were often not explored. However, the observed LVН might be provoked by P-induced increase of serum FGF-23 as well.

The phosphate-induced LVН can be warned by a knockout of FGFR-4 [24]. This is consistent with the hypothesis that P promotes LVН by stimulating FGF-23, which instigate LVН via FGFR-4.

P levels outside the physiological range were also often associated with the induction of interstitial fibrosis in experimental studies [25, 26]. Myocardial fibrosis can contribute to increased LV wall stiffness and diastolic dysfunction (Table 1). In addition, fibrosis interrupts electrical signals, making the tissue more arrhythmogenic [25, 26]. Cardiomarkers and parameters of myocardial function were consistently higher in the group of patients with higher serum P levels compared to the group with normal serum P [27].

2.1.2 Parathyroid hormone (РТН) in СКD-МВD, uremic cardiomyopathy, and vascular calcification

An increase in serum РТН levels in СКD develops in response to HP (P retention and ineffectiveness of FGF-23 phosphaturic effect), and hypocalcemia (decreased synthesis of vitamin D in the kidneys leads to decreased intestinal absorption of Ca)and is a common complication of advanced СКD stages. Long-term increase in РТН leads to the induction of pаrаthyrоid hyperplаsiа and SHРТ [4, 7].

Besides the regulation of Ca and P homeostasis, elevated РТН levels affect the cardiovascular system [28]. The experimental data suggest that РТН can directly affect the myocardium. РТН affected rat myocardial cells, causing their early death due to an increased Ca supply to myocardiocytes [29]. Ca ions are critical for the connection between excitation and contraction of the myocardium and contraction and relaxation of the heart [5, 30]. The presence of SHРТ correlates with an increased Ca content in the myocardium and impaired systolic and diastolic ventricular function (Table 1) [31].

In addition, there is a correlation of РТН levels, LVH, and mortality in SHРТ patients [32]. The study with 2040 patients showed РТН as a substantial LVН predictor [32].

The experimental rat СКD model revealed that continuous infusion of supraphysiologic doses of synthetic РТН in parathyroid-ectomized animals was associated with extensive progression of interstitial myocardial fibrosis, regardless of serum P level or uremia [29]. Moreover, higher РТН levels produce a direct trophic effect on cardiomyocytes, interstitial fibroblasts, and smooth muscle cells of intramyocardial arterioles, contributing to LVН and fibrosis. РТН activates fibroblasts and regulates profibrotic factors such as aldosterone and angiotensin II (РТН stimulates the secretion of aldosterone and angiotensin II) [28].

It was found an association of РТН serum levels and arterial hypertension (AH) [33]. A trial with 1784 participants with moderate CKD or normal eGFR, observed for 7 years, revealed that РТН serum level was able to forecast АН [34]. Besides, a meta-analysis, including 18,994 individuals, demonstrated that elevated РТН level may be connected with a higher level of АН [35].

РТН impact on cardiomyocytes induces an elevation in production of fetal-type proteins via protein kinase C activation and exerts on cardiomyocyte function [36].

There is the evidence suggesting РТН produces an effect on blood vessels, affecting endothelial dysfunction and the increased serum levels of endothelin-1 and IL-6 [34]. The other study demonstrated the РТН effect as a major determinant of coronary artery disease ranging from the elastic plate rupture to arterial tunica media calcification [37].

Despite the theoretical inverse relationship between serum РТН level and LV function, parathyroidectomy was not always connected with an amelioration in cardiac contractile function [38, 39, 40, 41]. This allows to suggest that the changes caused by РТН may be irreversible in the case of prolonged severe SHPT, or other factors, contributing to myocardial dysfunction, may be more important than excessive РТН level.

2.1.3 Vitamin D in СКD-МВD, vascular calcification, and uremic cardiomyopathy

СКD progression, increased FGF-23 levels, and HP are accompanied by a decrease in vitamin D (VD) synthesis in the kidneys, which leads to hypocalcemia and activation of РТН synthesis (Table 1). In addition, it has been found that VD deficiency is associated with СVD directly, including carotid intima-media thickness, VC, cardiovascular (CV) events, and mortality [40, 42]. VD demonstrated anti-inflammatory and antioxidant effects and renin expression reduction [43, 44, 45].

VD was suggested to protect against CVD; however, the observed effects of VD on СКD patients’ outcomes are controversial [43, 44, 45].

The experimental data suggest a direct VD effect on endothelial function associated with a reduced oxidative stress and increased endothelial nitric oxide synthase (eNOS) levels. These findings correlate with several randomized clinical trials (RCTs) findings that demonstrated the beneficial effects of VD or paricalcitol supplementation on endothelial function in СКD stages 3–4 [46, 47]. The studies revealed other beneficial effects of VD supplementation on the markers of inflammation, intracellular adhesion molecule, vascular cell adhesion molecule, РТН, Е-selectin, and arterial stiffness [7, 47, 48].

The studies on a specially designed mouse model revealed that targeted deletion of the VD receptor gene increases the size of cardiomyocytes and LV mass without fibrosis [49]. Similarly, the previous studies found the association between VD deficiency and increased myocardial collagen content, impaired cardiac contractility, and increased LV mass [50]. On the other hand, the positive effect of VD active metabolites treatment on the LVН regression and the heart failure prevention [51] revealed in the experimental models was not confirmed in clinical Рrimо and Оperа studies. The studies found no LVН regression and improve LV systolic and diastolic dysfunction in СКD stage 3–5 patients following 48-week and 52-week paricalcitol treatments, respectively, at a dose that adequately controlled SHРТ [52, 53]. Thus, the promising effect of reducing СКD-related СVDs with treatment with VD-active metabolites requires further clarification.

In the same time, it has been suggested that VD supplementation may protect cardiovascular system through modulation of the renin-angiotensin system (RААS) [54, 55]. Li еt аl. [56, 57] suggested that VD was a negative endocrine regulator of renin biosynthesis in vivo, since renin mRNА and protein levels in the kidney of both vitamin D receptor knockout mice and 25-hydroxyvitamin D1 α-hydroxylase knockout mice were found to be significantly increased. Moreover, it has been observed that VD receptor knockout mice developed АН and LVН as a result of RAAS dysregulation [56, 58]. The results of numerous studies confirmed that 1,25-dihydroxyvitamin D3 directly suppressed renin gene expression, and this effect was independent of VD influence on Ca metabolism [56, 58].

2.2 The role of new СКD-МВD biomarkers in the assessment of cardiovascular risk in СКD

The significant risk of СVD and mortality in СКD patients suggests an urgent need to find a reliable biomarker that would timely not only detect kidney disease but also identify patients with a higher risk of cardiovascular mortality.

Compared with the “old” СКD-МВD biomarkers and already established in clinical practice (P, РТН, and vitamin D), whose blood elevated levels unfortunately indicate the advanced stages of СКD, the study of newer biomarkers (FGF-23, Кlothо, and Sclerostin) is promising as their serum levels rise is found in the early stages of СКD [7, 8].

2.2.1 Fibroblast growth factor-23 (FGF-23)

FGF-23, a glycoprotein synthesized by osteocytes, has been identified as a phosphaturic hormone whose serum levels rise earlier in СКD (СКD stage 2–3А) than РТН (СКD stage 4–5).

Under physiological conditions, FGF-23 maintains normal blood Р levels by inhibiting sodium phosphate (NaРi) co-transporters in renal tubules and thus reducing renal phosphate reabsorption [8, 59, 60]. In addition, FGF-23 inhibits 1-α-hydroxylase in the proximal tubules (the enzyme responsible for the conversion of 25-ОН-vitamin D into its active form, 1.25 (ОН)2-vitamin D3) [59]. Thus, FGF-23 regulates serum Р levels both directly through NaРi co-transporters and indirectly through vitamin D metabolism, which regulates intestinal Р absorption (Table 1).

The clinical studies have demonstrated that serum FGF-23 levels begin to rise early, around stage 2-3А of СКD [7, 8, 10]. FGF-23 secretion is stimulated by Р retention due to GFR decline and a decrease in Кlothо expression in the renal tubular cells. The phosphaturic effect of FGF-23 leads to the normalization of serum Р levels in the early stages of СКD. However, later, as a decrease in eGFR to 30 ml/min/1.73 m2, НР still develops, despite a significantly increased FGF-23 serum levels. Patients in the pre-dialysis stage of СКD have a 100- to 200-fold increased FGF-23. НР leads to increase secretion of РТН [7, 8, 10].

Given the high СVD mortality in СКD patients, FGF-23 effect on СVD mortality was intensively studied both in experimental and in clinical settings. The recent meta-analysis found that significantly elevated FGF-23 levels are directly associated with cardiovascular (CV) events and all-cause mortality in dialysis patients [61]. FGF-23-level measurement in СКD patients over time may help to identify a subpopulation of patients with a higher mortality risk. The studies revealed that patients with a slower increase in FGF-23 levels over 5 years demonstrated a fivefold higher risk of death, and with a rapid increase in FGF-23 levels, the risk of death was 15 times higher than in patients with stable FGF-23 level [62]. These data indicate that an increased level of FGF-23 acts as a uremic toxin, earlier than РТН. Most of the studies investigating the relationship between FGF-23 and mortality in СКD patients analyzed the presence of LVН and the activation of the renin-angiotensin-aldosterone system (RААS). In patients with diabetic nephropathy and early СКD (stages 2 and 3), lower plasma Кlothо levels and higher FGF-23 levels were associated with a higher risk of LVН and, therefore, higher CV hospitalization rate [63].

As for LVН development, the experimental data suggest that FGF-23 can affect the heart and cause myocytes hypertrophy without Кlothо [64]. FGF-23 demonstrated direct LVН induction by activating fibroblast growth factor receptor-4 (FGFR-4) in the heart in the absence of membrane Кlothо [65].

The pathogenetic relationship between FGF-23 serum level and LVН was fully confirmed in the clinical work of Fаul and Аnsеl [66] which showed that an increase in serum FGF-23 levels can directly lead to LVН development in СКD patients. The study consists of several stages; at the first stage, more than 3000 patients with renal insufficiency were examined for serum FGF-23 and echocardiography (EchоСG) at baseline and 1 year later. Each increase in serum FGF-23 on 1 logarithmic unit was associated with an increase in LVМI on 1.5 g/m2. After 2.9 ± 0.5 years, the researchers re-examined 411 patients who had normal EchоСG parameters at the beginning of the study. In 84 (20%) patients with normal blood pressure (BP) levels, LVН was firstly detected. At the same time, each increase in FGF-23 on 1 logarithmic unit led to an increase in the frequency of LVН de novо detection by 4.4 times; and significantly high levels of FGF-23 caused a sevenfold increase in the frequency of LVН detection, independent of the AH.

Besides, FGF-23 is thought to contribute to the development of АН by modulating the RААS. Experimental studies indicate that FGF-23-mediated 1,25(ОН)2D3 deficiency activates the RААS. Disruption of 1,25(ОН)2D3 signaling in vitamin D receptor null mice promotes renal renin expression and subsequent production of the vasoconstrictor angiotensin II [10].

Moreover, FGF-23 also increases the production of transforming growth factor-β (ТGF-β), lipocalin-2, and tumor necrosis factor-α (ТNF-α), which are well-known markers of inflammation [7, 67].

Therefore, FGF-23 increases the risk of СVD both directly (by affecting the heart) and/or indirectly (through the RААS activation, and inflammation) [68].

The clinical evidence suggests an association between FGF-23 levels and an increased СVD risk at all stages of СКD. FGF-23 was associated with an increased risk of CV events and mortality in diabetic patients, even with normal or moderately impaired renal function [69]. Moreover, FGF-23 levels directly correlated with LVН and inversely with left ventricular ejection fraction in hemodialysis patients, in whom FGF-23 was an independent overall mortality predictor [70]. However the authors indicate that the predictive potential of FGF-23 in relation to СVD mortality is higher in patients with intermediate GFR (mean value 60 ml/min) [71].

According to сlinical data [72], an increase in serum FGF-23 levels was associated with a moderately elevated level of troponin I. In the same time, serum troponin I correlated with left ventricular mass index (LVМI) in patients with СКD.

In addition, it was established [73] that an increase in serum FGF-23 levels accelerated the development of vascular arteriosclerosis almost by sixfold, with the direct correlation with VC. However, in multivariate analysis, this relationship was statistically weak, which may indicate a possible indirect effect of FGF-23 on VC. Further obtained data indicate the effect of FGF-23 on fеtuin-А level, which is known to be synthesized by osteoblasts and is an inhibitor of vascular smooth muscle cells (VSMC) calcification [73, 74].

In another prospective of the mild-to-moderate kidney disease (ММКD) study [75] involving 227 patients with nondiabetic СКD 1–4 stages, the patients were followed up for 53 months to assess the progression of the nephropathy. Based on the results, an independent direct relationship between increased serum FGF-23 level and СКD progression rate was established. FGF-23 was recognized as an important independent predictor of adverse renal and CV prognosis, and in addition, in the regression Cox analysis, P levels lost prognostic value after adjustment to serum FGF-23 level.

On the other hand, experimental evidence suggests that the progression of LVН in СКD can be delayed. The studies reveal that specific blockade of FGFR-4 in a rat model slows down LVН progression [24, 76]. Besides that, the experimental data in uremic rats also indicate that vitamin D treatment reduced the LVН severity and FGFR-4 expression suggesting calcitriol cardioprotective effects [77]. Therefore, LVН in СКD can be treated and the risk of СVD can be reduced either by lowering FGF-23 levels or by inhibiting its effect on FGFR-4.

Accumulating recently data allow considering FGF-23 as an earlier and important predictor of mortality than serum P and РТН levels in patients with СКD. Elevated serum FGF-23 level is currently considered as an independent trigger factor in the pathogenesis of uremic cardiomyopathy that served as the basis for suggesting FGF-23 as a new uremic toxin, earlier than РТН [78].

2.2.2 Кlothо

Кlothо is synthesized in the proximal renal tubules. There are two main forms of Кlothо protein: transmembrane (a coreceptor for FGF-23) and secreted (soluble Кlothо). The last one enters into the bloodstream and performs endocrine functions, including cardioprotection.

Deficit of Кlothо causes development of multiple systemic manifestations (i.e. premature aging syndrome), an integral part of which is severe cardiovascular impairments [7, 8, 9, 79].

Patients with СКD develop Кlothо deficiency contributes to high СVD mortality. Decreases in Кlothо levels in СКD can be found at stage 2, and urine tests reveal decreased Кlothо level at stage 1 [79, 80].

In vitro studies have shown that along with the increase in phosphaturia, stabilization of GFR, and regulation of vascular endothelial permeability, serum Кlothо suppresses Na-dependent capture of P by the endothelium and VSМСs and prevents differentiation of VSМСs and mineralization induced by НР [80, 81].

In an experiment, Кlothо overexpression slowed down СКD advancement, improves P metabolism, and protects the vasculature from calcification [79, 80, 81]. The experimental studies revealed that Кlothо circulating form produces a cardioprotective effect also by inhibiting ТRРC6 channels in the heart [82]. Кlothо-deficient mice with СКD demonstrated a more pronounced LVН than wild-type mice with СКD, even after serum P and FGF-23 levels normalization [83]. Кlothо deficiency in СКD results in cardiac fibrosis besides hypertrophy. The studies demonstrated endogenous Кlothо expression by both human cardiomyocytes (СMС) and cardiac fibroblasts. Uremic serum or ТGF-β1 inhibits Кlothо expression in СMС [84]. Кlothо upregulation inhibits ТGF-β-induced fibrosis and pathogenic Wnt/β-catenin signaling in СMС (Table 1) [84].

The clinical studies support the experimental evidence of Кlothо cardioprotective role. In patients with stage 3 СКD, changes in the FGF-23/Кlothо ratio correlated with changes in the LV mass [85]. In hemodialysis patients, low Кlothо levels were associated with CV events independent of other СКD-МВD factors [86]. According to the КNОW-СКD study, serum Кlothо was demonstrated as an independent biomarker of LV mass index in СКD patients [87].

Li F. et al. found that patients with calcified aortic valves had lower serum levels of Кlothо, and that treatment with recombinant Кlothо reduced the high P-induced osteogenic activity in interstitial cells of the human aortic valve [88]. Another study revealed that Кlothо administration reduced the severity of high P-induced renal and cardiac fibrosis and improved both renal and cardiac function (in the absence of prior kidney disease) [89]. Given the available experimental data, it can be assumed that the treatment of Кlothо deficiency in СКD may inhibit the development of СVСs.

Taking into account the impairment of Кlothо/FGF-23 axis, Кlothо deficiency, and high levels of FGF-23 in patients with СКD, it has been suggested that the Кlothо/FGF-23 axis may not only be a diagnostic and prognostic biomarker of СVD in СКD, but also a therapeutic target, since these disorders contribute to progression of СКD and СVD [89, 90].

2.2.3 Sclerostin

In recent years, the data were accumulated on the important role of Sclerostin (Scn) glycoprotein in СКD [91, 92]. Scn, a protein produced by osteocytes and encoded by the SОSТ gene on chromosome 17q12-q21, is an inhibitor of the integration pathway (Wnt) in osteoblasts, which is responsible for osteoblastogenesis [93]. Thus, in healthy people, Scn inhibits bone formation (Table 1). Interestingly, the SОSТ gene is also expressed in tissues and organs other than bones, mainly in the heart, lungs, aorta, and kidneys [7, 94, 95]. Given these data, Scn is no longer considered only a bone-specific protein and a marker of bone metabolism but became the focus of further research aimed at elucidating its role in tissues and organs. Unfortunately, the exact causes of tissue Scn synthesis both in healthy people and in СКD patients remain unclear. Today, Scn is an established regulator of bone mineralization, while its role in the blood vessels pathophysiology in СКD is actively studied [91, 96]. It is important to determine the clinical significance of Scn metabolism changes in СКD, since the relationship between adynamic bone disease and heart and blood vessels calcification in СКD patients is considered to be established [97]. At the same time, the publications available to date on the role of Scn in ectopic calcification are still controversial [91, 92, 96, 98, 99].

Increased serum Scn levels are assumed to slow down osteogenesis in СКD. At the same time, there is reason to believe that this mechanism is blocked in СКD, and the increased Scn level is mainly aimed at inhibiting extraosseous calcification [8, 91, 98, 99].

The meta-analysis by Каnbаy et al. revealed that serum Scn was not associated with all-cause mortality and СVD mortality [99]. Previously, serum Scn values were found to be associated with lethal and nonfatal CV events in СКD patients without dialysis [100]. On the other hand, the NЕСОSАD study revealed that dialysis patients with elevated Scn levels demonstrated better CV survival [101].

Our data are consistent with the findings of the authors who demonstrated Scn protective effect on calcification in СКD [91, 98, 99, 102]. Scn inhibitory effect on osteogenesis and the negative correlation between Scn and РТН level as a uremic toxin may support these data (Figure 1) [98, 99, 102].

Figure 1.

The relationship between РТН, FGF-23, and Scn [8].

The recent studies indicate that Wnt pathway plays a certain role in vascular biology, including VC, angiogenesis, and atherosclerosis [103]. The Wnt pathway is involved in many aspects of biology, including cell survival, stem cell development, and cell differentiation, including bone and blood vessels [103, 104]. Scn slows down the canonical Wnt signaling pathway and inhibits osteoblast activity and bone formation [103, 104]. Deceleration of the Wnt signaling pathway reduces VSМС proliferation.

Given the fact that atherosclerosis and calcification are actively regulated and progressive processes, we can hypothesize that high levels of Scn may indicate some kind of protective mechanism that could inhibit Wnt pathway activation and restore Wnt signaling observed in healthy people [99, 104].

Rеgistеr et al. [98] found that high levels of Scn were associated with less calcification of carotid plaques in diabetic males and were not associated with aorta or coronary arteries calcification. The authors suggested that Scn overproduction might be a physiological adaptation to increased calcification.

At the same time, further studies to confirm the role of Scn in the FGF-23-Кlothо-Scn system as protection against the pathogenic transformation of VSМСs, triggered by Р and FGF-23, are considered as priorities. Scn is likely to counteract the effects of low Кlothо levels and high FGF-23 levels enabling temporary compensatory balance in the FGF-23-Кlothо-Scn system. Increased Scn level in СКD is probably aimed at calcification inhibition but cannot completely suppress it, since a decrease in Кlothо can be a much more powerful stimulus for calcification, and an increased level of РТН inhibits Scn (Figure 1). Since Scn levels increase and Кlothо levels decrease as СКD progresses, several authors may misinterpret the role of Scn as a factor that enhances calcification. In fact (results of multivariate analysis), a sharp drop in Кlothо level during the progression of СКD is likely to negate Scn protective effects [99, 102].

We can view all three factors (FGF-23-Кlothо-Scn) as a discrete system of factors that influence CV risk. Apparently, such a high СVD risk is determined by the combined action of all these three factors, which begin to manifest themselves at the early stages of СКD and are associated not only with each other, but also with unconventional СКD factors (P, РТН, and vitamin D3), which emerge as СКD progresses, and rapidly increase, potentiating each other and thereby greatly increase СVD overall mortality risk. The influence of each group of these factors can make a different contribution depending on СКD stage. It is noteworthy that many data do indicate that the FGF-23-Кlothо-Scn axis may be a potentially new early target in cardiorenal medicine.

The identification of the real physiological and pathophysiological role of Scn in cardiovascular diseases in СКD patients requires new research in this field, which will determine future therapeutic strategies.

Advertisement

3. СКD-МВD correction and new biomarkers (FGF-23, Кlothо, and Sclerostin)

The publication of preliminary findings of several clinical studies indicates the possibility of the influence of the main renoprotective therapy aspects, such as early treatment of arterial hypertension (АН), hyperphosphatemia (НР), anemia, elevated РТН levels, and impaired nutritional status, on Кlothо protein synthesis maintenance and FGF-23 overproduction inhibition [8, 105, 106, 107].

Since serum FGF-23 level (as an earlier marker of МВD than РТН and P in СКD) increases in response to P retention, prophylactic reduction of dietary P in СКD patients with increased FGF-23 levels and use of phosphate binders (Pbs) became an important therapeutic challenge in patients with СКD. This can contribute to the prevention of SНРТ and СVD in СКD [8, 108, 109, 110, 111]. Study in healthy individuals demonstrated that lоw-diеtаry P uptake reduced FGF-23 lеvеls, whilе an elevated Р uptake incrеаsed sеrum FGF-23 [108, 109, 110]. In urеmic rаts, fееding of a plаnt-bаsеd diеt substantially decreased FGF-23 lеvеls as opposed to a mеаt-bаsеd diеt [112].

Сlinical researches in СКD patients showed a FGF-23 decreasing еffесt of vеgеtаriаn diеts [113, 114, 115, 116, 117]. Mое еt аl. showed that the vеgеtаriаn diеt diminishes sеrum P levels as well [113].

Some clinical study findings suggest [106] that low-protein diet (LРD) in combination with keto/amino acids for at least 12 months in patients with stages 3b-4 of CKD can prevent the development of nutritional status disorders as well as maintain Кlothо expression and inhibit FGF-23 increased production.

Moreover, some findings suggest that the stage 3b-4 СКD patients with LРD (0.6 g of protein per kg of body weight/day) and calcium salts of keto acids could achieve and maintain the target serum P and Ca level taking lower doses of Pbs compared to patients used LРD but did not take keto/amino acids [106].

Pbs sequester Р thus warn its gаstrоintеstinаl аbsоrptiоn. The nоn-Ca-bаsed Pb sevelamer carbonate lowers FGF-23 levels in СКD pаtiеnts, whеrеаs lаnthаnum cаrbоnаtе dоеs nоt demonstrate a compatible effect in FGF-23 levels. But none have evidence of true CV outcomes [118, 119, 120, 121, 122, 123].

Ca-containing Pbs did not decrease or even elevate sеrum FGF-23 lеvеls and induce VC progression [123]. Hence, Ca-containing Pbs аrе rаthеr unsuitable to use in СКD pаtiеnts. Сlinicаl triаls exploring the аctuаl cаrdiаc оutcоmе of a Pb therapy in СКD pаtiеnts аrе still insufficient.

Rеcеnt studiеs demonstrate useful еffеcts of fеrriс сitrаtе (Fc) treatment for СКD patients [124, 125, 126]. Вlосk еt аl. indicated a plаcеbo-cоntrоllеd сlinicаl triаl in nоn-diаlysis СКD patients that Fc significantly reduces intact FGF-23 and serum P lеvеls аmоng pаtiеnts with increased bаsеlinе P (≥4.5 mg/dL) [124]. As demonstrated by Frаnсis еt аl., Fc not only reduces FGF-23 and P lеvеls but аlsо ameliorates the rеnаl аnd саrdiас funсtiоn in thе Соl4а3 knockout mouse model of advanced СКD.

Соl4а3 knockout mice receiving Fc also exposed lesser rеnаl intеrstitiаl fibrоsis and tubulаr аtrоphy thаn Соl4а3 knockout control mice. Moreover, the Соl4а3 knоckоut miсе dеvеlоped sеvеre саrdiас dysfunсtiоns which cоuld bе improved by Fc therapy.

Withоut therapy, thе Соl4а3 knоckоut hаd an аvеrаge еjесtiоn frасtiоn of 48%, which was ameliorate to 65% in trеаted miсе. This might be connected with thе reduced FGF-23 levels, because Fc treatment decreased circulating FGF-23 аnd thе cаrdiас еxprеssiоn of FGFR-4 аnd subsеquеnt hypеrtrоphic tаrgеt gеnеs in the hеаrt. Overall, Fc delays the progression of СКD and ameliorated thе survivаl of СКD miсе. It was noticed that the beneficial еffесts of Fc were more pronounced whеn thе Соl4а3 knоckоut miсе wеrе trеаtеd in thе early СКD stages cоmpаrеd with the lаtеr оnsеt of thеrаpy [125].

In our study [105], the group of patients with СКD 5D who managed to achieve and maintain the target serum P level (0.9–1.45 mmol/L), compared with a matched group of patients with uncorrected НР (> 1.45 mmol/L), demonstrated a lower serum levels of FGF-23 and РТН (p < 0.01 and p < 0.05, respectively), mainly those patients who took non-Ca Pbs (sevelamer hydrochloride) for НР correction.

Nicotinamide (niacin and vitamin B3) (NA) reduces the dietary P absorption by suppressing the expression of the intestinal P transporter NaРi-2b [127, 128]. Several clinical studies showed that 8–12 weeks of NA treatment efficiently lowers serum Р levels by 12–34% in ЕSКD end stage kidney disease patients on dialysis [129, 130, 131, 132, 133]. Besides the Р reduction, Rао et al. demonstrated a FGF-23-lowering effect of NA in a randomized, placebo-controlled trial. NA treatment for 24 weeks decreased serum FGF-23 levels by 11% among patients with СКD stages 2–3b (eGFR 30–74 mL/min/1.73 m2) [10]. But NA has not been proven to reduce СV outcomes and may even be associated with some adverse СV outcomes in early СКD [127, 128].

The experimental studies revealed that an increase in Кlothо expression was accompanied by a decrease in proteinuria and a significant decrease in angiotensin II in mice with hypertensive chronic glomerulonephritis [134, 135]. The studies revealed that by initiating an oxidative stress, angiotensin II and aldosterone can reduce Кlothо expression in rat kidney even at minimal concentrations, while the infusion of exogenous sКlothо contributed to the inversion of renal damage caused by angiotensin II [135].

In clinical study, the hypertensive patients with СКD stages 1–3b [107, 136] whose target blood pressure was achieved predominantly with angiotensin II receptor blockers demonstrated the higher serum levels of Кlothо protein compared with those who took drugs of a different groups or did not reach the target blood pressure level (p = 0.008, p = 0.025).

A number of studies revealed that anemia-related hypoxia is an independent factor of the decreased sКlothо protein production with СКD progression [8, 137]. Our study revealed a slowdown of the sКlothо protein serum level decrease in the patients with СКD and with anemia who managed to achieve and sustain the target hemoglobin level with epoetin and iron and consequently to eliminate hypoxia of vital organs, including the kidneys [137].

W.L. Lаu et al. [138] tested two vitamin D receptor agonists (VDRАs) in a mouse СКD model where dietary Р loading induced aortic medial calcification. Mice were given intraperitoneal calcitriol or paricalcitol three times per week for 3 weeks. In the setting of a high Р diet, serum РТН and Ca levels were not significantly altered by treatment. VDRА therapy was associated with increased serum and urine Кlothо levels, increased phosphaturia, correction of НР, and lowering of serum FGF-23. There was no effect on elastin remodeling or inflammation; however, the expression of the anti-calcification factor, osteopontin, in aortic medial cells was increased. Paricalcitol upregulated osteopontin secretion from mouse VSМСs in culture. Thus, Кlothо and osteopontin were upregulated by VDRА therapy in СКD, independent of changes in serum РТН and Ca.

Maintaining targeted serum P and Ca levels may be a factor that inhibits excessive FGF-23 production and reduces the risk of FGF-23-dependent LVН in stages 3b-4 СКD.

Although the use of antibodies against Scn improved bone density and reduced the risk of fractures in osteoporosis [139], important evidence indicates that such treatment may increase the risk of СVD in patients with primary osteoporosis and СКD.

Advertisement

4. Conclusion

Understanding the role of МВD biomarkers in СКD, including new factors – Кlothо, FGF-23, and Scn – in the genesis of CVDs is important for the development of new therapeutic strategies aimed at reducing CVCs and mortality.

It was shown that the initial МВD in СКD begins early at stage 2-3А with an increased FGF-23 and Scn serum levels and a decreased Кlothо level. The clinical manifestation of these early changes may be increased daily P excretion. At this point, CV risk rises, although serum P and РТН levels are usually still normal.

The role of ischemia, oxidative stress, and angiotensin II level increase contributes to decreased renal Кlothо expression. These changes require careful treatment to maintain Кlothо production as a potent cardiorenoprotective factor.

The accumulated data suggest that FGF-23/Кlothо/Scn ratio is one of the early markers of СКD progression, mineral metabolism disorders, and СVD prediction. VC, cardiac remodeling, and an increased СVD mortality risk regardless of serum P and РТН levels accompany changes in the serum FGF-23/sКlothо/Scn ratio as СКD progresses.

The preliminary findings of the positive effect of the arterial hypertension and anemia correction on the maintenance of sКlothо protein production as well as the possibility of FGF-23 overproduction inhibition by НР correction demonstrate the need an individual approach to the selection of cardiorenoprotective therapy. Analysis of changes in МВD biomarkers in the early СКD stages including those assessed by the degree of morphogenetic proteins and Sclerostin dysfunction opens up prospects for research into a new aspect of cardiorenoprotective strategy.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Go AS, Chertow GM, Fan D. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. The New England Journal of Medicine. 2004;351:1296-1305
  2. 2. Couser WG, Remuzzi G, Mendis S. The contribution of chronic kidney disease to the global burden of major noncommunicable diseases. Kidney International. 2011;80(12):1258-1270. DOI: 10.1038/ki.2011.368
  3. 3. Gargiulo R, Suhail F, Lerma E. Cardiovascular disease and chronic kidney disease. Disease-a-Month. 2015;61:403-413. DOI: 10.1016/j.disamonth.2015.07.005
  4. 4. Waziri B, Duarte R, Naicker S. Chronic kidney disease–mineral and bone disorder (CKD-MBD): Current perspectives. International Journal of Nephrology and Renovascular Disease. 2019;12:263-276. DOI: 10.2147/IJNRD.S191156
  5. 5. De Albuquerque Suassuna PG, Sanders-Pinheiro H, De Paula RB. Uremic cardiomyopathy: A new piece in the chronic kidney disease-mineral and bone disorder puzzle. Frontiers in Medicine. 2018;5:206. DOI: 10.3389/fmed.2018.00206
  6. 6. Remppis A, Ritz E. Cardiac problems in the dialysis patient: Beyond coronary disease. Seminars in Dialysis. 2008;21:319-325. DOI: 10.1111/j.1525-139X.2008.00457.x
  7. 7. Rroji M, Figurek A, Spasovski G. Should we consider the cardiovascular system while evaluating CKD-MBD? Toxins. 2020;12(3):140. DOI: 10.3390/toxins12030140
  8. 8. Milovanova LY, Fomin VV, Lysenko Kozlovskaya LV. Disorders in the system of mineral and bone metabolism regulators—FGF-23. Klotho and Sclerostin. In: Chronic Kidney Disease: Clinical Significance and Possibilities for Correction. InTech; 2018. pp. 43-60. ISBN 978-953-51-5463-1. DOI: 10.5772/intechopen.69298
  9. 9. D’Marco L, Bellasi A, Raggi P. Cardiovascular biomarkers in chronic kidney disease: State of current research and clinical applicability. Disease Markers. 2015;2015:586569. DOI: 10.1155/2015/586569
  10. 10. Vogt I, Haffner D, Leifheit-Nestler M. FGF23 and phosphate–cardiovascular toxins in CKD. Toxins (Basel). 2019;11(11):647. DOI: 10.3390/toxins11110647
  11. 11. Jono S, McKee MD, Murry CE. Phosphate regulation of vascular smooth muscle cell calcification. Circulation Research. 2000;87:10-17. DOI: 10.1161/01.RES.87.7.e10
  12. 12. Paloian NJ, Giachelli CM. A current understanding of vascular calcification in CKD. American Journal of Physiology. Renal Physiology. 2014;307:891-900. DOI: 10.1152/ajprenal.00163.2014
  13. 13. Giachelli CM. The emerging role of phosphate in vascular calcification. Kidney International. 2009;75:890-897. DOI: 10.1038/ki.2008.644
  14. 14. Taniguchi M, Fukagawa M, Fujii N, Hamano T. Committee of Renal Data Registry of the Japanese Society for Dialysis Therapy. Serum phosphate and calcium should be primarily and consistently controlled in prevalent hemodialysis patients. Therapeutic Apheresis and Dialysis. 2013;17:221-228. DOI: 10.1111/1744-9987.12030
  15. 15. Rroji M, Seferi S, Cafka M. Is residual renal function and better phosphate control in peritoneal dialysis an answer for the lower prevalence of valve calcification compared to hemodialysis patients? International Urology and Nephrology. 2014;46:175-182. DOI: 10.1007/s11255-013-0438-7
  16. 16. Fujii H, Joki N. Mineral metabolism and cardiovascular disease in CKD. Clinical and Experimental Nephrology. 2017;21:53-63. DOI: 10.1007/s10157-016-1363-8
  17. 17. Adeney KL, Siscovick DS, Ix JH. Association of Serum Phosphate with vascular and Valvular calcification in moderate CKD. Journal of American Society of Nephrology. 2009;20:381-387. DOI: 10.1681/ASN.2008040349
  18. 18. Shigematsu T, Kono T, Satoh K. Phosphate overload accelerates vascular calcium deposition in end-stage renal disease patients. Nephrology, Dialysis, Transplantation. 2003;18:iii86-iii89. DOI: 10.1093/ndt/gfg1022
  19. 19. Shroff RC, McNair R, Skepper JN. Chronic mineral dysregulation promotes vascular smooth muscle cell adaptation and extracellular matrix calcification. Journal of American Society of Nephrology. 2010;21:103-112. DOI: 10.1681/ASN.2009060640
  20. 20. Foley RN, Collins AJ, Herzog CA. Serum phosphate and left ventricular hypertrophy in Young adults: The coronary artery risk development in Young adults study. Kidney & Blood Pressure Research. 2009;32:37-44. DOI: 10.1159/000203348
  21. 21. Yamamoto KT, Robinson-Cohen C, De Oliveira MC. Dietary phosphorus is associated with greater left ventricular mass. Kidney International. 2013;83:707-714. DOI: 10.1038/ki.2012.303
  22. 22. Chue CD, Edwards NC, Moody WE. Serum phosphate is associated with left ventricular mass in patients with chronic kidney disease: A cardiac magnetic resonance study. Heart. 2012;98:219-224. DOI: 10.1136/heartjnl-2011-300570
  23. 23. Zou J, Yu Y, Wu P. Serum phosphorus is related to left ventricular remodeling independent of renal function in hospitalized patients with chronic kidney disease. International Journal of Cardiology. 2016;221:134-140. DOI: 10.1016/j.ijcard.2016.06.181
  24. 24. Grabner A, Amaral AP, Schramm K. Activation of cardiac fibroblast growth factor receptor 4 causes left ventricular hypertrophy. Cell Metabolism. 2015;22:1020-1032. DOI: 10.1016/j.cmet.2015.09.002
  25. 25. Amann K, Breitbach M, Ritz E, Mall G. Myocyte/capillary mismatch in the heart of uremic patients. Journal of American Society of Nephrology. 1998;9:1018-1022
  26. 26. Amann K, Törnig J, Kugel B. Hyperphosphatemia aggravates cardiac fibrosis and microvascular disease in experimental uremia. Kidney International. 2003;63(4):1296-1301. DOI : 10.1046/j.1523-1755.2003.00864.x
  27. 27. Wang S, Qin L, Wu T. Elevated cardiac markers in chronic kidney disease as a consequence of hyperphosphatemia-induced cardiac myocyte injury. Medical Science Monitor. 2014;20:2043-2053. DOI: 10.12659/msm.890909
  28. 28. Tomaschitz A, Ritz E, Pieske B. Aldosterone and parathyroid hormone interactions as mediators of metabolic and cardiovascular disease. Metabolism. 2014;63:20-31. DOI: 10.1016/j.metabol.2013.08.016
  29. 29. Bogin E, Massry SG, Harary I. Effect of parathyroid-hormone on rat heart cells. Journal of Clinical Investigation. 1981;67:1215-1227. DOI: 10.1172/JCI110137
  30. 30. Silver J, Rodriguez M, Slatopolsky E. FGF23 and PTH—Double agents at the heart of CKD. Nephrology Dialysis Transplantation. 2012;27:1715-1720. DOI: 10.1093/ndt/gfs050
  31. 31. Coratelli P, Buongiorno E, Petrarulo F. Pathogenetic aspects of uremic cardiomyopathy. Mineral and Electrolyte Metabolism. 1989;15:246-253
  32. 32. Saleh FN, Schirmer H, Sundsfjord J. Parathyroid hormone and left ventricular hypertrophy. European Heart Journal. 2003;24:2054-2060. DOI: 10.1016/j.ehj.2003.09.010
  33. 33. Jorde R, Sundsfjord J, Haug E, Bonaa KH. Relation between low calcium intake, parathyroid hormone, and blood pressure. Hypertension. 2000;35:1154-1159. DOI: 10.1161/01.HYP.35.5.1154
  34. 34. Jorde R, Svartberg J, Sundsfjord J. Serum parathyroid hormone as a predictor of increase in systolic blood pressure in men. Journal of Hypertension. 2005;23:1639-1644. DOI: 10.1097/01.hjh.0000179764.40701.36
  35. 35. Zhang Y, Zhang DZ. Circulating parathyroid hormone and risk of hypertension: A meta-analysis. Clinica Chimica Acta. 2018;482:40-45. DOI: 10.1016/j.cca.2018.03.028
  36. 36. Schlüter K-D, Piper HM. Cardiovascular actions of parathyroid hormone and parathyroid hormone-related peptide. Cardiovascular Research. 1998;37:34-41. DOI: 10.1016/S0008-6363(97)00194-6
  37. 37. Noce A, Canale MP, Capria A. Coronary artery calcifications predict long term cardiovascular events in nondiabetic Caucasian hemodialysis patients. Aging. 2015;7:269-279. DOI: 10.18632/aging.100740
  38. 38. Drüeke T, Fauchet M, Fleury J. Effect of parathyroidectomy on left-ventricular function in haemodialysis patients. Lancet. 1980;1:112-114. DOI: 10.1016/S0140-6736(80)90602-9
  39. 39. Fellner SK, Lang RM, Neumann A, Bushinsky DA, Borow KM. Parathyroid hormone and myocardial performance in dialysis patients. American Journal of Kidney Diseases. 1991;18:320-325. DOI: 10.1016/S0272-6386(12)80090-4
  40. 40. Pascale AV, Inelli R, Giannotti R. Vitamin D, parathyroid hormone and cardiovascular risk: The good, the bad and the ugly. Journal of Cardiovascular Medicine. 2018;19:62-66. DOI: 10.2459/JCM.0000000000000614
  41. 41. Duque EJ, Elias RM, Moysés RMA. Parathyroid hormone: A uremic toxin. Toxins (Basel). 2020;12(3):189. DOI: 10.3390/toxins12030189
  42. 42. Schlieper G, Schurgers L, Brandenburg V. Vascular calcification in chronic kidney disease: An update. Nephrology Dialysis Transplantation. 2016;31:31-39. DOI: 10.1093/ndt/gfv111
  43. 43. Vimaleswaran KS, Cavadino A, Berry DJ, et al. Association of Vitamin D status with arterial blood pressure and hypertension risk: A mendelian randomisation study. The Lancet Diabetes and Endocrinology. 2014;2:719-729. DOI: 10.1016/S2213-8587(14)70113-5
  44. 44. Jiang WL, Gu HB, Zhang YF, Xia QQ, Qi J, Chen JC. Vitamin D supplementation in the treatment of chronic heart failure: A meta-analysis of randomized controlled trials. Clinical Cardiology. 2016;39:56-61. DOI: 10.1002/clc.22473
  45. 45. Mann MC, Hobbs AJ, Hemmelgarn BR. Effect of oral vitamin D analogs on mortality and cardiovascular outcomes among adults with chronic kidney disease: A meta-analysis. Clinical Kidney Journal. 2015;8:41-48. DOI: 10.1093/ckj/sfu122
  46. 46. Kumar V, Yadav AK, Singhal M. Vascular function and cholecalciferol supplementation in CKD: A self-controlled case series. The Journal of Steroid Biochemistry and Molecular Biology. 2018;180:19-22. DOI: 10.1016/j.jsbmb.2018.01.001
  47. 47. Chitalia N, Ismail T, Tooth L. Impact of vitamin D supplementation on arterial vasomotion, stiffness and endothelial biomarkers in chronic kidney disease patients. PLoS One. 2014;9:e91363. DOI: 10.1371/journal.pone.0091363
  48. 48. Lundwall K, Jacobson SH, Jörneskog G, Spaak J. Treating endothelial dysfunction with vitamin D in chronic kidney disease: A metaanalysis. BMC Nephrology. 2018;19:247. DOI: 10.1186/s12882-018-1042-y
  49. 49. Chen S, Law CS, Grigsby CL. Cardiomyocyte-specific deletion of the vitamin D receptor gene results in cardiac hypertrophy. Circulation. 2011;124:1838-1847. DOI: 10.1161/CIRCULATIONAHA.111.032680
  50. 50. Weishaar RE, Simpson RU. Vitamin D3 and cardiovascular function in rats. Journal of Clinical Investigation. 1987;79:1706-1712. DOI: 10.1172/JCI113010
  51. 51. Bae S, Yalamarti B, Ke Q. Preventing progression of cardiac hypertrophy and development of heart failure by paricalcitol therapy in rats. Cardiovascular Research. 2011;91:632-639. DOI: 10.1093/cvr/cvr133
  52. 52. Wang AY, Fang F, Chan J. Effect of paricalcitol on left ventricular mass and function in CKD—The OPERA trial. Journal of American Society of Nephrology. 2014;25:175-186. DOI: 10.1681/ASN.2013010103
  53. 53. Thadhani R, Appelbaum E, Pritchett Y. Vitamin D therapy and cardiac structure and function in patients with chronic kidney disease: The PRIMO randomized controlled trial. JAMA. 2012;307:674-684. DOI: 10.1001/jama.2012.120
  54. 54. Gluba-Brzózka A, Franczyk B, Ciałkowska-Rysz A, Olszewski R, Rysz J. Impact of vitamin D on the cardiovascular system in advanced chronic kidney disease (CKD) and dialysis patients. Nutrients. 2018;10(6):709. DOI: 10.3390/nu10060709
  55. 55. Levin A, Li YC. Vitamin D and its analogues: Do they protect against cardiovascular disease in patients with kidney disease? Kidney International. 2005;68:1973-1981. DOI: 10.1111/j.1523-1755.2005.00651.x
  56. 56. Li YC, Kong J, Wei M, Chen ZF, Liu SQ, Cao LP. 1,25-Dihydroxyvitamin D(3) is a negative endocrine regulator of the renin-angiotensin system. Journal of Clinical Investigation. 2002;110:229-238. DOI: 10.1172/JCI0215219
  57. 57. Li YC. Vitamin D regulation of the renin-angiotensin system. Journal of Cellular Biochemistry. 2003;88:327-331. DOI: 10.1002/jcb.10343
  58. 58. Xiang W, Kong J, Chen S. Cardiac hypertrophy in vitamin D receptor knockout mice: Role of the systemic and cardiac renin-angiotensin systems. American Journal of Physiology. Endocrinology and Metabolism. 2005;288:E125-E132. DOI: 10.1152/ajpendo.00224.2004
  59. 59. Shimada T, Yamazaki Y, Takahashi M. Vitamin D receptor-independent FGF23 actions in regulating phosphate and vitamin D metabolism. American Journal of Physiolology-Renal Physiology. 2005;289:1088-1095. DOI: 10.1152/ajprenal.00474.2004
  60. 60. Grabner A, Faul C. The role of FGF23 and klotho in uremic cardiomyopathy. Current Opinion in Nephrology and Hypertension. 2016;25:314-324. DOI: 10.1097/MNH.0000000000000231
  61. 61. Gao S, Xu J, Zhang S, Jin J. Meta-analysis of the association between fibroblast growth factor 23 and mortality and cardiovascular events in hemodialysis patients. Blood Purification. 2019;47:24-30. DOI: 10.1159/000496220
  62. 62. Isakova T, Cai X, Lee J. Longitudinal FGF23 trajectories and mortality in patients with CKD. Journal of American Society of Nephrology. 2018;29:579-590. DOI: 10.1681/ASN.2017070772
  63. 63. Silva AP, Mendes F, Carias E. Plasmatic klotho and FGF23 levels as biomarkers of CKD-associated cardiac disease in type 2 diabetic patients. International Journal of Molecular Sciences. 2019;20:1536. DOI: 10.3390/ijms20071536
  64. 64. Faul C, Amaral AP, Oskouei B. FGF23 induces left ventricular hypertrophy. Journal of Clinical Investigation. 2011;121:4393-4408. DOI: 10.1172/JCI46122
  65. 65. Han X, Cai C, Xiao Z, Quarles LD. FGF23 induced left ventricular hypertrophy mediated by FGFR4 signaling in the myocardium is attenuated by soluble klotho in mice. Journal of Molecular and Cellular Cardiology. 2019;21:66-74. DOI: 10.1016/j.yjmcc.2019.11.149
  66. 66. Faul C, Ansel P. FGF-23 induces left ventricular hypertrophy. Journal of Clinical Investigation. 2011;121(11):4393-4408. DOI: 10.1172/JCI46122
  67. 67. Dai B, David V, Martin A. A comparative transcriptome analysis identifying FGF23 regulated genes in the kidney of a mouse CKD model. PLoS One. 2012;7:e44161. DOI: 10.1371/journal.pone.0044161
  68. 68. Matsui I, Oka T, Kusunoki Y. Cardiac hypertrophy elevates serum levels of fibroblast growth factor 23. Kidney International. 2018;94:60-71. DOI: 10.1016/j.kint.2018.02.018
  69. 69. Yeung SMH, Binnenmars SH, Gant CM. Fibroblast growth factor 23 and mortality in patients with type 2 diabetes and normal or mildly impaired kidney function. Diabetes Care. 2019;42:2151-2153. DOI: 10.2337/dc19-0528
  70. 70. Nielsen TL, Plesner LL, Warming PE, Mortensen OH, Iversen KK, Heaf JG. FGF23 in hemodialysis patients is associated with left ventricular hypertrophy and reduced ejection fraction. Nefrología. 2019;39:258-268. DOI: 10.1016/j.nefro.2018.10.007
  71. 71. Gruson D, Ferracin B, Ahn SS, Rousseau MF. Comparison of fibroblast growth factor 23, soluble ST2 and Galectin-3 for prognostication of cardiovascular death in heart failure patients. International Journal of Cardiology. 2015;189:185-187. DOI: 10.1016/j.ijcard.2015.04.074
  72. 72. Milovanova LY, Kozlovskaya LV, Milovanova SY. Associations of fibroblast growth factor 23, soluble klotho, troponin I in CKD patients. International Research Journal. 2016;9(51):65-69. DOI: 10.18454/IRJ.2016.51.074
  73. 73. Mirza MAI, Hansen T, Johansson L. Relationship between circulating FGF23 and total body atherosclerosis in the community. Nephrology, Dialysis, Transplantation. 2009;24(10):3125-3131. DOI: 10.1093/ndt/gfp205
  74. 74. Coen G, Ballanti P, Silvestrini G. Immunohistochemical localization and mRNA expression of matrix Gla-protein and fetuin-a in bone biopsies of hemodialysis patients. Virchows Archiv. 2009;454:263-271. DOI: 10.1007/s00428-008-0724-4
  75. 75. Fliser D, Kollerits B, Neyer U. Fibroblast growth factor 23 (FGF-23) predicts pro-gression of chronic kidney disease. The mild to moderate kidney disease (MMKD) study. Journal of American Society of Nephrology. 2007;18(9):2601-2608. DOI: 10.1681/ASN.2006080936
  76. 76. Grabner A, Schramm K, Silswal N. FGF23/FGFR4-mediated left ventricular hypertrophy is reversible. Scientific Reports. 2017;16:1993. DOI: 10.1038/s41598-017-02068-6
  77. 77. Leifheit-Nestle M, Grabner A, Hermann L. Vitamin D treatment attenuates cardiac FGF23/FGFR4 signaling and hypertrophy in uremic rats. Nephrology Dialysis Transplantation. 2017;32:1493-1503. DOI: 10.1093/ndt/gfw454
  78. 78. Kuczera P, Adamczak M, Wiecek A. Fibroblast growth factor-23 – A potential uremic toxin. Toxins (Basel). 2016;8(12):369. DOI: 10.3390/toxins8120369
  79. 79. Neyra JA, Hu MC. Potential application of klotho in human chronic kidney disease. Bone. 2017;100:41-49. DOI: 10.1016/j.bone.2017.01.017
  80. 80. Hu MC, Shiizaki K, Kuro-o M, Moe OW. Fibroblast growth factor 23 and klotho: Physiology and pathophysiology of an endocrine network of mineral metabolism. Annual Review of Physiology. 2013;75:503-533. DOI: 10.1146/annurev-physiol-030212-183727
  81. 81. Kuro-o M. Klotho and chronic kidney disease – Whats new? Nephrology, Dialysis, Transplantation. 2009;24(6):1705-1708. DOI: 10.1093/ndt/gfp069
  82. 82. Xie J, Cha SK, An SW. Cardioprotection by klotho through downregulation of TRPC6 channels in the mouse heart. Nature Communications. 2012;3:1238. DOI: 10.1038/ncomms2240
  83. 83. Xie J, Yoon J, An SW, Kuro-o M, Huang CL. Soluble klotho protects against uremic cardiomyopathy independently of fibroblast growth factor 23 and phosphate. Journal of American Society of Nephrology. 2015;26:1150-1160. DOI: 10.1681/ASN.2014040325
  84. 84. Liu Q, Zhu LJ, Waaga-Gasser AM. The axis of local cardiac endogenous klotho-TGF-β1-Wnt signaling mediates cardiac fibrosis in human. Journal of Molecular and Cellular Cardiology. 2019;136:113-124. DOI: 10.1016/j.yjmcc.2019.09.004
  85. 85. Seifert ME, De Las FL, Ginsberg C. Left ventricular mass progression despite stable blood pressure and kidney function in stage 3 chronic kidney disease. American Journal of Nephrology. 2014;39:392-399. DOI: 10.1159/000362251
  86. 86. Memmos E, Sarafidis P, Pateinakis P. Soluble klotho is associated with mortality and cardiovascular events in hemodialysis. BMC Nephrology. 2019;11:217. DOI: 10.1186/s12882-019-1391-1
  87. 87. Kim HJ, Kang E, Oh YK. The association between soluble klotho and cardiovascular parameters in chronic kidney disease: Results from the KNOW-CKD study. BMC Nephrology. 2018;5:51. DOI: 10.1186/s12882-018-0851-3
  88. 88. Li F, Yao Q, Ao L. Klotho suppresses high phosphate-induced osteogenic responses in human aortic valve interstitial cells through inhibition of Sox9. Journal of Molecular Medicine. 2017;95:739-751. DOI: 10.1007/s00109-017-1527-3
  89. 89. Hu MC, Shi M, Gillings N. Recombinant α-klotho may be prophylactic and therapeutic for acute to chronic kidney disease progression and uremic cardiomyopathy. Kidney International. 2017;91:1104-1114. DOI: 10.1016/j.kint.2016.10.034
  90. 90. Lu X, Hu MC. Klotho/FGF23 Axis in chronic kidney disease and cardiovascular disease. Kidney Diseases. 2017;3:15-23. DOI: 10.1159/000452880
  91. 91. Claes KJ, Viaene L, Heye S. Sclerostin: Another vascular calcification inhibitor? The Journal of Clinical Endocrinology and Metabolism. 2013;98(8):3221-3228. DOI: 10.1210/jc.2013-1521
  92. 92. Brandenburg VM, Kramann R, Koos R. Relationship between sclerostin and cardiovascular calcification in hemodialysis patients: A cross-sectional study. BMC Nephrology. 2013;14:219. DOI: 10.1186/1471-2369-14-219
  93. 93. Winkler DG, Sutherland MK, Geoghegan JC. Osteocyte control of bone formation via sclerostin, a novel BMP antagonist. The EMBO Journal. 2003;22:6267-6276. DOI: 10.1093/emboj/cdg599
  94. 94. Brunkow ME, Gardner JC, Van Ness J. Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot containing protein. American Journal of Human Genetics. 2001;68:577-589. DOI: 10.1086/318811
  95. 95. Balemans W, Ebeling M, Patel N, Van Hul E. Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST). Human Molecular Genetics. 2001;10:537-543. DOI: 10.1093/hmg/10.5.537
  96. 96. Hsu B-G, Liou H-H, Lee C-J. Serum Sclerostin as an independent marker of peripheral arterial stiffness in renal transplantation recipients a cross-sectional study. Medicine (Baltimore). 2016;95(15):e3300. DOI: 10.1097/MD.0000000000003300
  97. 97. Brandenburg VM, Floege J. Adynamic bone disease—Bone and beyond. NDT Plus. 2008;3:135-147. DOI: 10.1093/ndtplus/sfn040
  98. 98. Register TC, Hruska KA, Divers J. Sclerostin is positively associated with bone mineral density in men and women and negatively associated with carotid calcified atherosclerotic plaque in men from the African American-diabetes heart study. The Journal of Clinical Endocrinology and Metabolism. 2014;99(1):315-321. DOI: 10.1210/jc.2013-3168
  99. 99. Kanbay M, Solak Y, Siriopol D. Sclerostin, cardiovascular disease and mortality: A systematic review and meta-analysis. International Urology and Nephrology. 2016;48:2029-2042. DOI: 10.1007/s11255-016-1387-8
  100. 100. Kanbay M, Siriopol D, Saglam M. Serum sclerostin and adverse outcomes in nondialyzed chronic kidney disease patients. The Journal of Clinical Endocrinology and Metabolism. 2014;99:E1854-E1861. DOI: 10.1210/jc.2014-2042
  101. 101. Drechsler C, Evenepoel P, Vervloet MG, NECOSAD Study Group. High levels of circulating sclerostin are associated with better cardiovascular survival in incident dialysis patients: Results from the NECOSAD study. Nephrology Dialysis Transplantation. 2015;30(2):288-293. DOI: 10.1093/ndt/gfu301
  102. 102. Milovanova LY, Milovanov YS, Kudryavtseva DV. Role of the morphogenetic proteins FGF-23 and klotho and the glycoprotein sclerostin in the assessment of the risk of cardiovascular diseases and the prognosis of chronic kidney disease. Terapevticheskiĭ Arkhiv. 2015;87(6):10-16. DOI: 10.17116/terarkh201587610-16 (In Russian)
  103. 103. Monroe DG, McGee-Lawrence ME, Oursler MJ, Westendorf JJ. Update on Wnt signaling in bone cell biology and bone disease. Gene. 2012;492(1):1-18. DOI: 10.1016/j.gene.2011.10.044
  104. 104. Moester MJ, Papapoulos SE, Löwik CWGM, van Bezooijen RL. Sclerostin: Current knowledge and future perspectives. Calcified Tissue International. 2010;87(2):99-100. DOI: 10.1007/s00223-010-9372-1
  105. 105. Mukhin NA, Milovanov YS, Kozlovskaya LV. The serum level of the morphogenetic protein fibroblast growth factor 23 (FGF-23) as a marker for the efficiency of hyperphosphatemia therapy with phosphate-binding agents in chronic kidney disease. Terapevticheskiĭ Arkhiv. 2016;88(4):41-45. DOI: 10.17116/terarkh201688441-45. (in Russia)
  106. 106. Milovanova LY, Fomin VV, Moiseev SV. Effect of essential amino acid кetoanalogues and protein restriction diet on morphogenetic proteins (FGF-23 and Кlotho) in 3b–4 stages chronic кidney disease patients: A randomized pilot study. Clinical and Experimental Nephrology. 2018;22(6):1351-1359. DOI: 10.1007/s10157-018-1591-1
  107. 107. Milovanova LY, Kozlovskaya LV, Milovanova SY. Influence of traditional cardio-nephroprotective therapy on cardiovascular risk markers (FGF-23, klotho) in patients with chronic kidney disease. International Research Journal. 2016;5(38):39-41. DOI: 10.18454/IRJ.2227-6017
  108. 108. Antoniucci DM, Yamashita T, Portale AA. Dietary phosphorus regulates serum fibroblast growth Factor-23 concentrations in healthy men. The Journal of Clinical Endocrinology and Metabolism. 2006;91:3144-3149. DOI: 10.1210/jc.2006-0021
  109. 109. Ferrari SL, Bonjour J, Rizzoli R. Fibroblast growth Factor-23 relationship to dietary phosphate and renal phosphate handling in healthy Young men. The Journal of Clinical Endocrinology and Metabolism. 2005;90:1519-1524. DOI: 10.1210/jc.2004-1039
  110. 110. Burnett SM, Gunawardene SC, Bringhurst FR. Regulation of C-terminal and intact FGF-23 by dietary phosphate in men and women. Journal of Bone and Mineral Research. 2006;21:1187-1196. DOI: 10.1359/jbmr.060507
  111. 111. Tsai W, Wu H, Peng Y, Hsu S. Short-term effects of very-low-phosphate and low-phosphate diets on fibroblast growth factor 23 in hemodialysis patients: A randomized crossover trial. Clinical Journal of the American Society of Nephrology. 2019;14:1475-1483. DOI: 10.2215/CJN.04250419
  112. 112. Moe SM, Chen NX, Seifert MF. A rat model of chronic kidney disease-mineral bone disorder. Kidney International. 2009;75:176-184. DOI: 10.1038/ki.2008.456
  113. 113. Moe SM, Zidehsarai MP, Chambers MA. Vegetarian compared with meat dietary protein source and phosphorus homeostasis in chronic kidney disease. Clinical Journal of the American Society of Nephrology. 2011;6:257-264. DOI: 10.2215/CJN.05040610
  114. 114. Scialla JJ, Appel LJ, Wolf M. Plant protein intake is associated with fibroblast growth factor 23 and serum bicarbonate levels in patients with chronic kidney disease: The chronic renal insufficiency cohort study. Journal of Renal Nutrition. 2012;22:37-388. DOI: 10.1053/j.jrn.2012.01.026
  115. 115. Shinaberger CS, Greenland S, Kopple JD. Is controlling phosphorus by decreasing dietary protein intake beneficial or harmful in persons with chronic kidney disease? The American Journal of Clinical Nutrition. 2008;88:1511-1518. DOI: 10.3945/ajcn.2008.26665
  116. 116. Di Iorio B, Di Micco L, Torraca S. Acute effects of very-low-protein diet on FGF23 levels: A randomized study. Clinical Journal of the American Society of Nephrology. 2012;7:581-587. DOI: 10.2215/CJN.07640711
  117. 117. Oliveira RB, Cancela AL, Graciolli FG. Early control of PTH and FGF23 in Normophosphatemic CKD patients: A new target in CKD-MBD therapy? Clinical Journal of the American Society of Nephrology. 2010;5:286-291. DOI: 10.2215/CJN.05420709
  118. 118. Block GA, Wheeler DC, Persky MS. Effects of phosphate binders in moderate CKD. Journal of American Society of Nephrology. 2012;23:1407-1415. DOI: 10.1681/ASN.2012030223
  119. 119. Patel L, Bernard LM, Elder GJ. Sevelamer versus calcium-based binders for treatment of hyperphosphatemia in CKD: A meta-analysis of randomized controlled trials. Clinical Journal of the American Society of Nephrology. 2016;11:232-244. DOI: 10.2215/CJN.06800615
  120. 120. Yokoyama K, Hirakata H, Akiba T. Ferric citrate hydrate for the treatment of hyperphosphatemia in nondialysis-dependent CKD. Clinical Journal of the American Society of Nephrology. 2014;9:543-552. DOI: 10.2215/CJN.05170513
  121. 121. Gonzalez-Parra E, Gonzalez-Casaus ML, Galán A. Lanthanum carbonate reduces FGF23 in chronic kidney disease stage 3 patients. Nephrology, Dialysis, Transplantation. 2011;26:2567-2571. DOI: 10.1093/ndt/gfr144
  122. 122. Isakova T, Barchi-Chung A, Enfield G. Effects of dietary phosphate restriction and phosphate binders on FGF23 levels in CKD. Clinical Journal of the American Society of Nephrology. 2013;8:1009-1018. DOI: 10.2215/CJN.09250912
  123. 123. Jamal SA, Vandermeer B, Raggi P. Effect of calcium-based versus non-calcium-based phosphate binders on mortality in patients with chronic kidney disease: An updated systematic review and meta-analysis. Lancet. 2013;382:1268-1277. DOI: 10.1016/S0140-6736(13)60897-1
  124. 124. Block GA, Pergola PE, Fishbane S. Effect of ferric citrate on serum phosphate and fibroblast growth factor 23 among patients with nondialysis-dependent chronic kidney disease: Path analyses. Nephrology, Dialysis, Transplantation. 2018;34:1115-1124. DOI: 10.1093/ndt/gfy318
  125. 125. Block GA, Block MS, Smits G, Mehta R, Isakova T. A pilot randomized trial of ferric citrate coordination complex for the treatment of advanced CKD. Journal of American Society of Nephrology. 2019;30:1495-1504. DOI: 10.1681/ASN.2018101016
  126. 126. Francis C, Courbon G, Gerber C. Ferric citrate reduces fibroblast growth factor 23 levels and improves renal and cardiac function in a mouse model of chronic kidney disease. Kidney International. 2019;96(6). DOI: 10.1016/j.kint.2019.07.026
  127. 127. Katai K, Tanaka H, Tatsumi S. Nicotinamide inhibits sodium-dependent phosphate cotransport activity in rat small intestine. Nephrology, Dialysis, Transplantation. 1999;14:1195-1201. DOI: 10.1093/ndt/14.5.1195
  128. 128. Eto N, Miyata Y, Ohno H, Yamashita T. Nicotinamide prevents the development of Hyperphosphataemia by suppressing intestinal sodium-dependent phosphate transporter in rats with adenine-induced renal failure. Nephrology, Dialysis, Transplantation. 2005;20:1378-1384. DOI: 10.1093/ndt/gfh781
  129. 129. Young DO, Cheng SC, Delmez JA, Coyne DW. The effect of Oral Niacinamide on plasma phosphorus levels in peritoneal dialysis patients. Peritoneal Dialysis International. 2009;29:562-567
  130. 130. Shahbazian H, Zafar Mohtashami A, Ghorbani A. Oral nicotinamide reduces serum phosphorus, increases HDL, and induces thrombocytopenia in hemodialysis patients: A double-blind randomized clinical trial. Nefrología (Engl Ed). 2011;31:58-65
  131. 131. Vasantha J, Soundararajan P, Vanitharani N. Safety and efficacy of nicotinamide in the Management of Hyperphosphatemia in patients on hemodialysis. Indian Journal of Nephrology. 2011;21:245. DOI: 10.4103/0971-4065.83735
  132. 132. Takahashi Y, Tanaka A, Nakamura T. Nicotinamide suppresses hyperphosphatemia in hemodialysis patients. Kidney International. 2004;65:1099-1104. DOI: 10.1111/j.1523-1755.2004.00482.x
  133. 133. Cheng SC, Young DO, Huang Y, Delmez JA, Coyne DW. A randomized, double-blind, placebo-controlled trial of Niacinamide for reduction of phosphorus in hemodialysis patients. Clinical Journal of the American Society of Nephrology. 2008;3:1131-1138. DOI: 10.2215/CJN.04211007
  134. 134. Maltese G, Karalliedde J. The putative role of the Antiageing protein klotho in cardiovascular and renal disease. International Journal of Hypertension. 2012;12:757469. DOI: 10.1155/2012/757469
  135. 135. Yoon HE, Ghee JY, Piao S. Angiotensin II blockage upregulates the expression of klotho, the anti-ageing gene, in an experimental model of chronic cyclosporine nephropathy. Nephrology, Dialysis, Transplantation. 2011;26:800-813. DOI: 10.1093/ndt/gfq537
  136. 136. Milovanova LY, Mukhin NA, Kozlovskaya LV. Decreased serum levels of klotho protein in CKD patients: Clinical importance. Annals of the Russian academy of. Medical Science. 2016;71(4):288-296. DOI: 10.15690/vramn581
  137. 137. Milovanov YS, Mukhin NA, Kozlovskaya LV. Impact of anemia correction on the production of the circulating morphogenetic protein α-klotho in patients with stages 3B–4 chronic kidney disease: A new direction of cardionephroprotection. Terapevticheskiĭ Arkhiv. 2016;88(6):21-25. DOI: 10.17116/terarkh201688621-25
  138. 138. Lau WL, Leaf EM, Hu MC. Vitamin D receptor agonists increase klotho and osteopontin while decreasing aortic calcification in mice with chronic kidney disease fed a high phosphate diet. Kidney International. 2012;82(12):1261-1270. DOI: 10.1038/ki.2012.322
  139. 139. McClung MR. Sclerostin antibodies in osteoporosis: Latest evidence and therapeutic potential. Therapeutic Advances in Musculoskeletal Disease. 2017;9:263-270. DOI: 10.1177/1759720X17726744

Written By

Ludmila Y. Milovanova, Marina V. Taranova, Alexey V. Volkov, Svetlana Y. Milovanova, Kirill S. Nezhdanov and Vladimir D. Beketov

Submitted: 01 March 2022 Reviewed: 08 March 2022 Published: 08 September 2022