Next Article in Journal
Limited Proteolysis of Cyclooxygenase-2 Enhances Cell Proliferation
Next Article in Special Issue
Roles of Histone Acetylation Modifiers and Other Epigenetic Regulators in Vascular Calcification
Previous Article in Journal
Transcriptomic Changes Related to Cellular Processes with Particular Emphasis on Cell Activation in Lysosomal Storage Diseases from the Group of Mucopolysaccharidoses
Previous Article in Special Issue
Vascular Calcification—New Insights into Its Mechanism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Sclerostin in Bone and Ectopic Calcification

Lab of Pathophysiology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, 2610 Antwerp, Belgium
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(9), 3199; https://doi.org/10.3390/ijms21093199
Submission received: 15 April 2020 / Revised: 27 April 2020 / Accepted: 29 April 2020 / Published: 30 April 2020
(This article belongs to the Special Issue Mechanisms of Vascular Calcification)

Abstract

:
Sclerostin, a 22-kDa glycoprotein that is mainly secreted by the osteocytes, is a soluble inhibitor of canonical Wnt signaling. Therefore, when present at increased concentrations, it leads to an increased bone resorption and decreased bone formation. Serum sclerostin levels are known to be increased in the elderly and in patients with chronic kidney disease. In these patient populations, there is a high incidence of ectopic cardiovascular calcification. These calcifications are strongly associated with cardiovascular morbidity and mortality. Although data are still controversial, it is likely that there is a link between ectopic calcification and serum sclerostin levels. The main question, however, remains whether sclerostin exerts either a protective or deleterious role in the ectopic calcification process.

1. Discovery of Sclerostin

In the 1950s, sclerosteosis and van Buchem disease, two rare autosomal recessive disorders characterized by massive and progressive bone overgrowth, were described for the first time [1,2]. Since both diseases are clinically and radiographically very similar, it was speculated that these two conditions result from mutations in the same gene [3]. Genome-wide linkage analysis on both sclerosteosis and van Buchem disease patients indeed revealed co-localization of the disease gene loci in the chromosomal region 17q12-q21 [4,5]. In sclerosteosis patients, extensive sequence analysis in this chromosomal region identified loss-of-function mutations in a previously unknown gene, now called the SOST gene [6]. In van Buchem disease patients, however, no disease-causing mutations were found within this gene [6,7]. By further sequencing downstream of the SOST gene, a 52kb deletion (containing a regulatory element) was identified, which affects the transcription of the SOST gene in bone [8,9,10].
The SOST gene product ‘sclerostin’ is a 22-kDa protein, and is a well-known negative regulator of bone formation. Although generally viewed as an osteocyte-specific protein, other tissues such as the kidney, liver, bone marrow, lung, heart and pancreas also express SOST mRNA [6,7]. In contrast to sclerosteosis patients, in which functional sclerostin is completely absent, van Buchem disease patients have a reduced sclerostin expression compared to healthy controls [6,11]. This is in line with the milder clinical phenotype that is observed in van Buchem disease patients, compared to sclerosteosis patients.

2. The Role of Sclerostin in Physiological Calcification

The canonical Wnt/β-catenin signaling pathway, in addition to its function during embryogenesis [12,13], also plays a crucial role in adult tissue homeostasis by regulating the maintenance and differentiation of stem cells. In particular, this signaling cascade also exerts an important regulatory pathway in the differentiation of mesenchymal stem cells towards the osteoblast-lineage.
Beta-catenin is the central regulatory player in the canonical Wnt signaling. Activation of this signaling cascade, by binding of the Wnt ligands to the Frizzled (Fz) receptor and Low-density Lipoprotein Receptor-related Protein 5/6 (LRP5/6) co-receptors, leads to inhibition of the β-catenin degradation complex. In this way, β-catenin can accumulate in the cytoplasm, and subsequently be translocated into the nucleus. In the nucleus, β-catenin functions as a coactivator of the transcription factors T-cell factor (TCF) and Lymphoid Enhancer-binding factor (LEF), thereby modifying gene transcription. It has been shown that the Wnt/β-catenin signaling cascade downregulates adipogenic differentiation by inhibiting the expression of Peroxisome Proliferator-Activated Receptor gamma (PPARγ) and CCAAT/Enhancer Binding Protein alpha (C/EBPα), both important adipogenic regulators, while stimulating Runt-related transcription factor 2 (Runx2) and Osterix, well-known inducers of osteogenesis [14,15]. The canonical Wnt signaling also stimulates osteoblast maturation and viability of osteoblasts and osteocytes. These cells then increase their production of osteoprotegerin (OPG) (a decoy receptor of Receptor Activator of Nuclear Factor Kappa-Β Ligand (RANKL)), by which osteoclast formation is inhibited.
To prevent excessive bone formation, several antagonists are produced amongst which is sclerostin. Mechanical unloading [16], low levels of serum parathyroid hormone (PTH) [17,18] and estrogen deficiency [19] trigger sclerostin production. As already mentioned above, in the bone, sclerostin is mainly produced by the osteocytes, the cells that reside within the bone matrix and comprise between 90%–95% of all bone cells. After its secretion, sclerostin will be anchored to the LRP4 receptor on the osteoblast membrane, by which sclerostin is retained in the bone compartment [20]. Sclerostin can also bind to LRP5/6, leading to receptor internalization and/or reduced availability of these co-receptors to Wnt ligands, which results in inhibition of the canonical Wnt signaling. This leads to (Figure 1):
 I. 
Inhibition of proliferation and differentiation of osteoprogenitor/pre-osteoblastic cells, as well as decreased activation of mature osteoblasts
Osteoblasts are derived from mesenchymal stems cells, which are multipotent progenitor cells that are able to differentiate into a variety of cell types (including osteoblasts, chondrocytes, adipocytes, smooth muscle cells [21] and endothelial cells [22]). Depending on the specific activation of signaling pathways (such as Wnt/β-catenin signaling) and transcription factors (such as Runx2 and osterix), mesenchymal cells will commit to the osteoblastic lineage. Inhibition of the canonical Wnt signaling by sclerostin therefore directly prevents the development of new osteoblasts. However, Thouverey and Caverzasio found that sclerostin not only functions by inhibiting canonical Wnt signaling, but also activates platelet-derived growth factor receptor signaling to inhibit osteoblast differentiation [23]. Sclerostin also inhibits the activity of mature osteoblasts, since osteocalcin, procollagen type 1 N-terminal Propeptide (P1NP) and bone-specific alkaline phosphatase (BsAP), all produced by the osteoblast and therefore considered indicators of osteoblastic activity, were increased after administration of romosozumab, an antibody directed against sclerostin [24].
 II. 
Decreased mineralization
Mineralization of newly formed bone is a dynamic process in which Small Integrin-Binding Ligand N-linked Glycoproteins (SIBLINGS), such as Matrix Extracellular Phosphoglycoprotein (MEPE), are involved [25]. A key characteristic of MEPE, and several other SIBLING proteins, is the presence of an Acidic Serine Aspartate-Rich MEPE-associated (ASARM) motif [26]. When cleaved by cathepsin B, the ASARM motif inhibits mineralization and phosphate uptake [27]. Cleavage of this ASARM motif can be prevented by Phosphate-regulating neutral Endopeptidase (PHEX), which binds to full-length MEPE [28,29]. Regulation of mineralization is therefore determined by the PHEX/MEPE ratio. Sclerostin is involved in the regulation of this PHEX/MEPE axis, by inducing the expression of ASARM-peptides (anti-mineralization) and downregulating PHEX (pro-mineralization) [30,31].
 III. 
Increased apoptosis of the osteogenic cells
Experiments in Sost knockout (KO) mice demonstrated decreased apoptosis of osteoblasts and osteocytes, while osteoblast activity was increased in these animals compared to wild type (WT) mice [32]. These results were confirmed by Chandra et al., showing that in a mouse model of radiation damage, the inhibition of sclerostin protected against apoptosis of osteoblasts [33]. Additionally, in human osteoblastic cells, sclerostin induced apoptosis by activating caspases 1, 3, 4 and 7, as well increasing the expression of the pro-apoptotic factor Bax [34].
 IV. 
Maintenance of bone lining cells in the quiescent state
Bone lining cells are found covering the bone surface. They are considered to be derived from previously active osteoblasts that did not undergo apoptosis or differentiated into osteocytes [35]. During normal physiology, active bone lining cells play a crucial role as coordinators of bone formation and resorption [36,37]. Regulation of the activity state of the bone lining cells—either remaining in the quiescent state or being reactivated to osteoblasts—at least in part, seems to be controlled by sclerostin. One study, performed in ovariectomized rats and cynomolgus monkeys, demonstrated that anti-sclerostin antibody treatment strongly reduced quiescent bone surfaces, whilst increasing bone surfaces actively involved in mineralization [38]. Another study in which mice were administered with an anti-sclerostin antibody, showed reactivation of the bone lining cells, as indicated by the increase in size and expression of osteocalcin, a marker of osteoblastic bone formation [39]. These results demonstrate that reactivation of bone lining cells could explain the rapid increase in osteoblast numbers on previously quiescent bone surfaces [39].
 V. 
Regulation of osteocyte maturation and osteocytic osteolysis
Immature osteocytes, which are located in the osteoid or in close proximity of the bone surface, still share many morphological features with the osteoblast [40]. However, when the osteocytes becomes deeper embedded in the mineralized bone matrix, they change their morphology to become mature osteocytes [40]. This process of osteocyte maturation is triggered by matrix mineralization [40].
As discussed above, sclerostin is an inhibitor of matrix mineralization and therefore, could also be involved in the regulation of osteocyte maturation. This was demonstrated by Atkins et al., who found that, after treating human primary osteoblast cultures with sclerostin, cells increased their E11 expression (pre-osteocyte marker), while decreasing dentin matrix acidic phosphoprotein 1 (DMP1) and SOST expression (mature osteocyte markers) [30].
In the bone, sclerostin is produced by the mature osteocytes, where its expression is regulated by a wide variety of factors, including local cytokines [41], hormones such as PTH [18] and estrogen [19], and mechanical loading [42]. Under unloading conditions, sclerostin is produced by the osteocytes in order to regulate re-shaping of the osteocytic lacunae by ‘osteocytic osteolysis’ [43]. The proteins that are upregulated by sclerostin correspond to those produced by the osteoclasts, including carbonic anhydrase 2 [43], cathepsin K [43,44], tartrate-resistant acid phosphatase [43,44] and C-terminal collagen telopeptide [31]. This process of osteocytic osteolysis has important effects on bone physiology, not only by releasing calcium from the bone matrix, which is shown to be crucial during lactation [44], but also by affecting mechano-sensation and bone turnover (reviewed by Tsourdi et al. [45]).
 VI. 
Stimulation of bone resorption
Research has shown that the canonical Wnt/β-catenin signaling is a critical regulator of osteoclastogenesis [46,47,48]. During the inhibition of canonical Wnt signaling, the expression of OPG is decreased, thereby increasing the RANKL/OPG ratio, and, thus, bone resorption [49]. Wei et al. showed that, in response to RANKL, β-catenin in osteoclast precursors is downregulated, which is needed to allow the differentiation of osteoclast precursors towards mature osteoclasts [46]. Furthermore, Weivoda et al. demonstrated that osteoclast lineage cells express canonical Wnt receptors [48], indicating that sclerostin might have direct effects on osteoclast formation and maturation.

3. The Role of Sclerostin in Ectopic Calcification

Ectopic calcification (i.e., inappropriate mineralization, which can occur in various soft tissues) is normally prevented by the presence of local and systemic calcification inhibitors. The absence of one or more of these calcification inhibitors allows the development of calcifications that are typically composed of calcium phosphate salts, such as hydroxyapatite. Although ectopic calcifications can develop in various parts of the body, cardiovascular tissues, skin, kidney and tendons seem particularly prone [50]. The deposition of calcification in tissues, which do not calcify under normal physiological conditions, may lead to serious adverse clinical effects.

3.1. Vascular Calcification

Calcification of the vascular tree is associated with aging, however, is accelerated in the presence of diabetes, chronic kidney disease (CKD), osteoporosis, dyslipidemia and certain genetic diseases. In these patient populations, vascular calcification importantly contributes to increased morbidity and mortality. Vascular calcification is an actively regulated and complex process that shares many similarities with bone development and metabolism. During the vascular calcification process, vascular smooth muscle cells lose their smooth muscle cell markers (e.g., α smooth muscle actin and smooth muscle protein 22 α), and obtain the characteristics of bone-like cells—a process called osteochondrogenic transdifferentiation. This goes along with loss of mineralization inhibitors, the formation of calcifying matrix vesicles, degradation of the extracellular matrix and vascular smooth muscle cell (VSMC) death [51,52,53]. Since the canonical Wnt signaling is known to be a crucial regulator of bone turnover, this signaling pathway could therefore also be involved in vascular calcification.
Two different types of vascular calcification can be distinguished depending on their location in the vascular wall: intima calcification, which is associated with atherosclerosis, and media calcification, or Mönkeberg’s sclerosis.
Intima calcification, which frequently affects the aorta and large elastic arteries, is characterized by patchy calcifications in the vicinity of lipid/cholesterol deposits. Plaque growth, which goes along with progressing intima calcification, causes a narrowing of the arterial lumen, leading to ischemia in the downstream organs and tissues. Acute rupture of the plaque’s fibrous cap, on the other hand, can result in thrombosis or infarction. To which extent the presence of calcification influences plaque stability is still controversial (reviewed by Barrett et al. [54]). Experimental evidence demonstrated the involvement of the canonical Wnt signaling in atherosclerosis, more specifically its role in endothelial dysfunction [55,56,57,58,59], macrophage activation [60,61,62,63] and VSMC proliferation and migration [64,65]. Considering the involvement of the canonical Wnt signaling in the development and progression of atherosclerosis, a possible role for sclerostin could be reserved. Research by Leto et al. identified sclerostin expression in atherosclerostic plaques in patients with and without type 2 diabetes [66]. Furthermore, in type 2 diabetes patients, there is an association between the presence of atherosclerotic disease and serum sclerostin levels [67]. Since Krishna et al. found that sclerostin decreases the expression of genes involved in matrix degradation and calcification, and thereby inhibits atherosclerosis [68], it is likely that sclerostin could function as an inhibitor of intimal vascular calcification.
Vascular media calcification, also named arteriosclerosis, or Mönckebergs’ sclerosis, is characterized by concentric calcifications in the tunica media of large elastic arteries, medium-sized visceral and renal arteries, as well as in small transitional arteries [69,70,71]. The principal consequences are increased wall thickness, arterial stiffening and development of left ventricular hypertrophy. This type of calcification, although frequently observed with aging in the general population, is strongly pronounced in patients with CKD, diabetes mellitus and other metabolic disorders. In CKD patients, elevated calcium and phosphate levels exert direct effects on VSMCs in order to stimulate vascular calcification. This is done by promoting osteochondrogenic transdifferentiation of VSMCs, VSMC apoptosis, matrix vesicle release, loss of calcification inhibitors and degradation of the extracellular matrix. Similar to intimal calcification, the canonical Wnt signaling pathway is also activated in vascular media calcification, as demonstrated by several studies [72,73,74,75,76,77,78,79,80,81]. Its activation is further regulated by numerous factors, as described in Figure 2. Most of the factors that activate the canonical Wnt signaling act by directly stimulating β-catenin expression and/or accumulation in the nucleus [73,78,82,83,84]. Other mechanisms, such as upregulation of Wnt ligands [77,85] and decreased expression of Wnt inhibitors [85], however, are also involved. Activation of the Wnt/β-catenin signaling, in turn, promotes vascular calcification by different mechanisms, amongst which are: (1) the modulation of Runx2 gene expression, which stimulates osteochondrogenic transdifferentiation of VSMCs [86]; (2) the induction of matrix metalloproteinases [79]; and (3) the upregulation of RANKL expression [81]. Given the important clinical consequences of vascular media calcification, many researchers have been investigating the inhibitors of the canonical Wnt signaling in the context of vascular media calcification. One of the mechanisms to inhibit activation of this signaling cascade is by preventing Wnt ligands to bind to their receptors. This can be achieved either by: (1) binding of secreted Frizzled-related proteins (sFRP) to Wnt ligands, which makes it impossible for Wnt ligands to bind to the Fz receptor [76],; or possibly (2) also by binding of sclerostin to the LRP4/5/6 receptors, which makes these receptors unavailable for Wnt-ligands [87,88]. Other mechanisms that have been described to inhibit vascular calcification by inactivating the Wnt/β-catenin signaling include magnesium [89] (and angiotensin II, which stimulates magnesium influx [90]), Klotho [91], PPARγ agonists [92,93], Collagen XIV [94], microRNAs [75,95,96], PTH receptor activation [97], Sirtuin1 [98,99], the anti-diabetic drug gemigliptin [100] and Ginkgo Biloba extract [88]. Data regarding the role of calcitriol and its analog paricalcitol in the activation of Wnt/β-catenin signaling and the vascular calcification process are conflicting. It is likely that their effects on vascular calcification development are dose-dependent [101,102].
The regulating factor that during the last years received the most attention, with regard to vascular calcification and Wnt-signaling, is sclerostin. Most research investigating the role of sclerostin in vascular media calcification is performed in the setting of CKD. In this patient population, the rapidly progressive development of vascular calcification is associated with significant morbidity and mortality. Compared to healthy individuals, end-stage renal disease (ESRD) patients have serum sclerostin levels that are three to four times higher [103,104]. This, however, does not seem to be attributed to reduced clearing by the kidneys; on the contrary, it was demonstrated that the elimination of sclerostin by the kidneys increases with declining kidney function [105]. A plausible explanation for these increased circulating sclerostin levels is an enhanced production of sclerostin. In this context, the osteocytes in the bone, which are the main producers of sclerostin, are to be considered first. One study in juvenile cystic kidney (jck) mice, a genetic model that develops progressive renal failure, showed that, in these mice, the number of osteocytes that express sclerostin increases during the very early stages of the disease compared to WT mice [106]. However, sclerostin expression returns to normal during the further course of the disease [106]. Since deterioration of the kidney function goes along with the development of vascular media calcification, transdifferentiated VSMCs could also be responsible for the observed increase in serum sclerostin levels. Although some studies did not find sclerostin to be expressed in calcified arteries [107], several others detected sclerostin mRNA and protein in calcified vessels [87,108,109]. It was suggested that ‘vascular’ sclerostin can contribute to increased serum levels in a non-CKD model for vascular calcification. The study, by De Maré et al., showed that serum sclerostin levels were increased, along with an increased expression of sclerostin in the vasculature, but without changes in the number of sclerostin-producing osteocytes [87]. Also, in calciphylaxis, a rare type of ectopic mineralization in which the calcifications are located in the medial layer of cutaneous arterioles, sclerostin is expressed in the calcified tissue and increased serum levels are observed [110,111].
Whether circulating sclerostin levels could function as a marker for the degree of vascular calcification remains unclear. Some studies find no association [112], while others find a positive [107,113,114,115,116,117,118] or negative correlation between circulating sclerostin levels and the extent of vascular calcification [119,120]. Several elements could have contributed to these conflicting results. Studies investigated the presence of vascular calcification at different anatomical locations, and used different statistical analyses (by adjusting for different potential confounding factors). In addition, differences in the time period between administration of enoxaparin (or other low molecular weight heparins that are used as anticoagulants) and blood collection could also be important, since enoxaparin stimulates the release of sclerostin into the circulation [121]. This also implies that the heterogeneity of study populations—i.e., patients at different CKD-stages, with or without dialysis treatment, whether or not they receive low molecular weight heparins—contributes to the observed inconsistency. Lastly, it is known that there are large discrepancies between sclerostin assays [122,123,124]. The antibodies that are used in the distinct assays bind different epitopes, therefore, some antibodies will capture only the intact sclerostin molecule, whilst others might also bind to sclerostin fragments.
The exact consequences of increased serum sclerostin levels in CKD patients are not unequivocally recognized. Studies investigating the association between serum sclerostin and (cardiovascular) mortality reported conflicting results, ranging from no association [118,125,126,127] to a positive one [128], as well as a negative association [119,129,130,131,132]. These inconsistent results could again be attributed to the use of different immunoassays to determine the serum sclerostin concentration. Moreover, the advanced/terminal condition of many CKD patients further complicates these kind of survival studies.
Conflicting results in the association between circulating sclerostin levels and severity of calcification/cardiovascular mortality could also be linked to sclerostin’s primary role, i.e., effects at the level of the bone. In addition to its potential effects on cardiovascular mortality, by altering the vascular calcification process, increased serum sclerostin levels can also have consequences at the level of the bone. In CKD patients, but also in osteoporosis patients, ectopic vascular calcification is frequently accompanied by a reduced bone mineral density and disturbed bone turnover. This seemingly contradictory association is called the calcification paradox [133]. During this pathological process, the regulation of several molecules and pathways involved in calcium and phosphate homeostasis (FGF23, PTH, Vit D, Klotho) is disturbed, leading to renal osteodystrophy/osteoporotic bone loss on the one hand, and ectopic calcification on the other hand. Also sclerostin might be involved in this process, since it is hypothesized that sclerostin produced in the vasculature can spill over into the circulation, via which it reaches the bone compartment, where it inhibits bone formation/mineralization/turnover [87,134] (Figure 3), thereby inhibiting the bone’s buffering capacity for calcium and phosphate, and making it available for deposition in the vessel wall.

3.2. Aortic Valve Calcification

Compared to patients with a normal renal function, there is an increased prevalence and accelerated progression of aortic valve calcification (AVC) in ESRD patients [135]. One study suggested the involvement of the canonical Wnt/β-catenin-signaling pathway in the differentiation of aortic valve interstitial cells into osteoblast-like cells [136]. A limited number of studies investigated the role of sclerostin in aortic valve calcification. One study demonstrated that the expression of sclerostin, which, in the aortic valve is located adjacent to the areas of calcification, was strongly associated with AVC in hemodialysis patients [137]. The same research group also compared the sclerostin expression in aortic valves, as well as serum levels in patients with and without aortic valve calcification [138]. Higher serum sclerostin levels were observed in patients with aortic valve calcification. Moreover, in these patients, sclerostin expression in the aortic valves was increased, as demonstrated by immunohistochemistry and gene expression analysis [138]. Lastly, Ji et al. demonstrated that serum sclerostin levels increase gradually, along with renal deterioration in CKD stage 3–5 patients, and that sclerostin can be an independent risk factor for aortic valve calcification in this patient population [139].

3.3. Genetic Disorders Characterized by Ectopic Calcification

Diffuse ectopic calcifications, located in all kinds of soft tissues, such as the arteries, myocardium, joints, liver and brain, are often the result of mutations in genes encoding for/or involved in the production of calcification inhibitors (MGP [140], pyrophosphate [141,142,143]), or proteins involved in phosphate regulation [144,145,146]. Data regarding the role of sclerostin during this ectopic calcification process are very limited. However, in a mouse model for primary familial brain calcification, it was demonstrated that sclerostin is expressed in vessel-associated calcifications in the brain [147].

3.4. Cancer-Associated Calcifications

Micro-calcifications are mainly observed in primary breast cancer [148], but can also be present in other types of cancer, such as prostate, thyroid and lung cancer [149,150,151]. Often, bone-related proteins are identified in these mineralized foci, indicating that this process might be similar to the physiological bone formation process [148]. To our knowledge there is currently no research investigating the role of sclerostin in this process.

3.5. Intracranial Calcification

Intracranial calcification refers to mineral depositions that are located within the brain parenchyma or the blood vessel wall [152,153]. This causes neuronal death and gliosis, resulting in a progressive deterioration of cognitive and motor skills, including dementia, Parkinsonism, psychosis, mood swings, dystonia and spastic paralysis [153]. Calcifications are mostly observed in the basal ganglia, but also in the thalamus, cerebellum and subcortical white matter [154]. The etiology behind this ectopic calcification process can be genetic predisposition, but also inflammatory and metabolic disorders [153]. Aberrant canonical Wnt signaling, has been shown to be involved in the pathogenesis of adamantinomatous craniopharyngiomas [155]. In these patients, gene expression analysis showed an increased expression of the canonical Wnt inhibitors, sFRP1 and Dickkopf-related protein 3 (DKK3) [156]. Further research is needed to determine the role of sclerostin in this disorder, and more generally in intracranial calcification.

4. Prospects

The role of sclerostin in ectopic calcification clearly needs to be investigated further, especially since an anti-sclerostin antibody, Romosozumab (EVENITY®), was approved last year for medical use in Japan, South Korea, Australia, Canada, US and the European Union [157]. This antibody binds to sclerostin, thereby preventing the interaction between sclerostin and LRP4/5/6 co-receptors. In osteoporosis patients, this monoclonal antibody successfully increased bone mineral density by inducing bone formation and decreasing bone resorption [24]. However, notwithstanding its clear benefits to bone health, there are concerns regarding the cardiovascular safety [158].
The ARCH study in postmenopausal women, compared weekly treatment with alendronate for 24 months versus monthly injection with Romosozumab for 12 months, followed by 12 months of alendronate treatment. Despite being superior in preventing fractures, treatment with Romosozumab resulted in a higher incidence of cardiovascular adverse events during the first year, compared to alendronate treatment [159]. These observations might point to a protective function of sclerostin in vascular calcification. As described above, studies demonstrated up-regulated expression of sclerostin in calcified vessels, and, although the function of sclerostin in the vasculature is not fully understood [87,108,109], this upregulation of sclerostin is hypothesized to be a negative feedback mechanism to prevent further calcification, which is also the case in bones [87,134]. In addition to the reduced hydroxyapatite formation by inhibiting canonical Wnt signaling, sclerostin might also prevent further calcification by stimulating dedifferentiated VSMCs to obtain adipocyte-like characteristics. Evidence for the involvement of sclerostin in adipogenesis comes from several in vitro [160,161] and in vivo studies [161,162,163]. Additionally, in the setting of ectopic vascular calcification, proteomic analysis demonstrated the upregulation of adipocyte-related proteins [87]. Amongst these proteins might be PPARγ, an important inducer of adipogenesis and known to be upregulated by sclerostin [160]. Furthermore, studies have shown that PPARγ activation is able to counteract vascular calcification [164,165].
Although preclinical studies did not detect any soft tissue mineralization due to the anti-sclerostin antibody treatment, it needs to be mentioned that radiography is not the most sensitive method to detect vascular calcification [166,167]. Additionally, in sclerosteosis and Van Buchem disease patients, no impairment of cardiac or vascular function has been reported. However, studies have shown that in order to compensate for the loss of sclerostin in sclerosteosis and Van Buchem disease patients, as well as in Sost KO mice, expression of other Wnt inhibitors such as Dickkopf-related protein 1 (DKK1) are upregulated [168,169]. Overexpression of DKK1 is not able to prevent excessive bone formation, however, it might be sufficient to prevent ectopic calcification. Furthermore, due to the high bone formation rate in these patients, in combination with reduced osteoclastic activity, most of the calcium and phosphate will be stored in the bone compartment, and will therefore not be available for the development of ectopic calcification. Another explanation for the (seemingly) increased incidence of cardiovascular events due to Romosozumab treatment, is the comparison with alendronate. This latter drug has potential cardioprotective effects [170], however, published results are contradictory [171,172]. Given the ambiguity regarding the cardiovascular risk when treated with Romosozumab, the prescribing information included a warning stating that “Romosozumab may increase the risk of myocardial infarction, stroke and cardiovascular death” [173]. Until the role of sclerostin in ectopic calcification has not been fully clarified, it is advised to cautiously outweigh benefits and risk in patients with cardiovascular risk factors.

Author Contributions

Drafted and revised the manuscript, A.D.M.; Revised the manuscript, P.C.D. and A.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Fund of Scientific Research-Flanders (FWO), grant number 1182417N and 1182419N.

Acknowledgments

A.D.M. is a fellow of the Fund of Scientific Research-Flanders (FWO).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

SOSTgene encoding sclerostin
WntWingless-related integration site
FzFrizzled
LRP5/6Low-density Lipoprotein Receptor-related Protein 5/6
TCFT-cell factor
LEFLymphoid Enhancer-binding factor
PPARγPeroxisome Proliferator-Activated Receptor γ
C/EBPαCCAAT/Enhancer-Binding Protein α
Runx2Runt-related transcription factor 2
OPGOsteoprotegerin
RANKLReceptor Activator of Nuclear Factor Kappa-Β ligand
PTHParathyroid hormone
LRP4Low-density Lipoprotein Receptor-related Protein 4
P1NPProcollagen type 1 N-terminal Propeptide
BsAPBone-specific Alkaline Phosphatase
SIBLINGSSmall Integrin Binding Ligand N-Glycoproteins
MEPEMatrix Extracellular Phosphoglycoprotein
ASARMAcidic Serine Aspartate-Rich MEPE-associated motif
PHEXPhosphate-regulating neutral Endopeptidase, X-linked
KOKnockout
WTWild type
BaxBcl2- associated X protein
DMP1Dentin matrix acidic phosphoprotein 1
CKDChronic kidney disease
VSMCVascular smooth muscle cell
sFRPsecreted Frizzled-related protein
ESRDEnd-stage renal disease
JckJuvenile cystic kidney
FGF23Fibroblast growth factor 23
MGPMatrix γ-carboxyglutamic acid
AVCAortic valve calcification
DKK3Dickkopf-related protein 3
DKK1Dickkopf-related protein 1

References

  1. Truswell, A.S. Osteopetrosis with syndactyly; a morphological variant of Albers-Schonberg’s disease. J. Bone Joint Surg. Br. 1958, 40, 209–218. [Google Scholar]
  2. Buchem, V. Hyperostosis corticalis generalisata familiaris (van Buchem’s disease). Acta Radiol. 1955, 44, 109–120. [Google Scholar] [CrossRef]
  3. Beighton, P.; Barnard, A.; Hamersma, H.; van der Wouden, A. The syndromic status of sclerosteosis and van Buchem disease. Clin. Genet. 1984, 25, 175–181. [Google Scholar] [CrossRef] [PubMed]
  4. Van Hul, W.; Balemans, W.; Van Hul, E.; Dikkers, F.G.; Obee, H.; Stokroos, R.J.; Hildering, P.; Vanhoenacker, F.; Van Camp, G.; Willems, P.J. Van Buchem disease (hyperostosis corticalis generalisata) maps to chromosome 17q12-q21. Am. J. Hum. Genet. 1998, 62, 391–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Balemans, W.; Van Den Ende, J.; Freire Paes-Alves, A.; Dikkers, F.G.; Willems, P.J.; Vanhoenacker, F.; de Almeida-Melo, N.; Alves, C.F.; Stratakis, C.A.; Hill, S.C.; et al. Localization of the gene for sclerosteosis to the van Buchem disease-gene region on chromosome 17q12-q21. Am. J. Hum. Genet. 1999, 64, 1661–1669. [Google Scholar] [CrossRef] [Green Version]
  6. Balemans, W.; Ebeling, M.; Patel, N.; Van Hul, E.; Olson, P.; Dioszegi, M.; Lacza, C.; Wuyts, W.; Van Den Ende, J.; Willems, P.; et al. Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST). Hum. Mol. Genet. 2001, 10, 537–543. [Google Scholar] [CrossRef] [Green Version]
  7. Brunkow, M.E.; Gardner, J.C.; Van Ness, J.; Paeper, B.W.; Kovacevich, B.R.; Proll, S.; Skonier, J.E.; Zhao, L.; Sabo, P.J.; Fu, Y.; et al. Bone dysplasia sclerosteosis results from loss of the SOST gene product, a novel cystine knot-containing protein. Am. J. Hum. Genet. 2001, 68, 577–589. [Google Scholar] [CrossRef] [Green Version]
  8. Balemans, W.; Patel, N.; Ebeling, M.; Van Hul, E.; Wuyts, W.; Lacza, C.; Dioszegi, M.; Dikkers, F.G.; Hildering, P.; Willems, P.J.; et al. Identification of a 52 kb deletion downstream of the SOST gene in patients with van Buchem disease. J. Med. Genet. 2002, 39, 91–97. [Google Scholar] [CrossRef]
  9. Staehling-Hampton, K.; Proll, S.; Paeper, B.W.; Zhao, L.; Charmley, P.; Brown, A.; Gardner, J.C.; Galas, D.; Schatzman, R.C.; Beighton, P.; et al. A 52-kb deletion in the SOST-MEOX1 intergenic region on 17q12-q21 is associated with van Buchem disease in the Dutch population. Am. J. Med. Genet. 2002, 110, 144–152. [Google Scholar] [CrossRef]
  10. Loots, G.G.; Kneissel, M.; Keller, H.; Baptist, M.; Chang, J.; Collette, N.M.; Ovcharenko, D.; Plajzer-Frick, I.; Rubin, E.M. Genomic deletion of a long-range bone enhancer misregulates sclerostin in Van Buchem disease. Genome Res. 2005, 15, 928–935. [Google Scholar] [CrossRef] [Green Version]
  11. van Lierop, A.H.; Hamdy, N.A.; van Egmond, M.E.; Bakker, E.; Dikkers, F.G.; Papapoulos, S.E. Van Buchem disease: Clinical, biochemical, and densitometric features of patients and disease carriers. J. Bone Miner. Res. 2013, 28, 848–854. [Google Scholar] [CrossRef] [PubMed]
  12. Tanaka, S.S.; Kojima, Y.; Yamaguchi, Y.L.; Nishinakamura, R.; Tam, P.P. Impact of WNT signaling on tissue lineage differentiation in the early mouse embryo. Dev. Growth Differ. 2011, 53, 843–856. [Google Scholar] [CrossRef] [PubMed]
  13. Nusse, R.; Varmus, H. Three decades of Wnts: A personal perspective on how a scientific field developed. Embo J. 2012, 31, 2670–2684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kang, S.; Bennett, C.N.; Gerin, I.; Rapp, L.A.; Hankenson, K.D.; MacDougald, O.A. Wnt signaling stimulates osteoblastogenesis of mesenchymal precursors by suppressing CCAAT/enhancer-binding protein alpha and peroxisome proliferator-activated receptor gamma. J. Biol. Chem. 2007, 282, 14515–14524. [Google Scholar] [CrossRef]
  15. Bennett, C.N.; Longo, K.A.; Wright, W.S.; Suva, L.J.; Lane, T.F.; Hankenson, K.D.; MacDougald, O.A. Regulation of osteoblastogenesis and bone mass by Wnt10b. Proc. Natl Acad Sci USA 2005, 102, 3324–3329. [Google Scholar] [CrossRef] [Green Version]
  16. Robling, A.G.; Niziolek, P.J.; Baldridge, L.A.; Condon, K.W.; Allen, M.R.; Alam, I.; Mantila, S.M.; Gluhak-Heinrich, J.; Bellido, T.M.; Harris, S.E.; et al. Mechanical stimulation of bone in vivo reduces osteocyte expression of Sost/sclerostin. J. Biol. Chem. 2008, 283, 5866–5875. [Google Scholar] [CrossRef] [Green Version]
  17. Bellido, T.; Ali, A.A.; Gubrij, I.; Plotkin, L.I.; Fu, Q.; O’Brien, C.A.; Manolagas, S.C.; Jilka, R.L. Chronic elevation of parathyroid hormone in mice reduces expression of sclerostin by osteocytes: A novel mechanism for hormonal control of osteoblastogenesis. Endocrinology 2005, 146, 4577–4583. [Google Scholar] [CrossRef]
  18. Keller, H.; Kneissel, M. SOST is a target gene for PTH in bone. Bone 2005, 37, 148–158. [Google Scholar] [CrossRef]
  19. Fujita, K.; Roforth, M.M.; Demaray, S.; McGregor, U.; Kirmani, S.; McCready, L.K.; Peterson, J.M.; Drake, M.T.; Monroe, D.G.; Khosla, S. Effects of Estrogen on Bone mRNA Levels of Sclerostin and Other Genes Relevant to Bone Metabolism in Postmenopausal Women. J. Clin. Endocr. Metab. 2014, 99, E81–E88. [Google Scholar] [CrossRef]
  20. Boudin, E.; Yorgan, T.; Fijalkowski, I.; Sonntag, S.; Steenackers, E.; Hendrickx, G.; Peeters, S.; De Mare, A.; Vervaet, B.; Verhulst, A.; et al. The Lrp4 R1170Q Homozygous Knock-In Mouse Recapitulates the Bone Phenotype of Sclerosteosis in Humans. J. Bone Miner. Res. 2017, 32, 1739–1749. [Google Scholar] [CrossRef] [Green Version]
  21. Gong, Z.; Niklason, L.E. Small-diameter human vessel wall engineered from bone marrow-derived mesenchymal stem cells (hMSCs). FASEB J. 2008, 22, 1635–1648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Oswald, J.; Boxberger, S.; Jorgensen, B.; Feldmann, S.; Ehninger, G.; Bornhauser, M.; Werner, C. Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem Cells 2004, 22, 377–384. [Google Scholar] [CrossRef] [PubMed]
  23. Thouverey, C.; Caverzasio, J. Sclerostin inhibits osteoblast differentiation without affecting BMP2/SMAD1/5 or Wnt3a/beta-catenin signaling but through activation of platelet-derived growth factor receptor signaling in vitro. Bonekey Rep. 2015, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Padhi, D.; Jang, G.; Stouch, B.; Fang, L.; Posvar, E. Single-dose, placebo-controlled, randomized study of AMG 785, a sclerostin monoclonal antibody. J. Bone Miner. Res. 2011, 26, 19–26. [Google Scholar] [CrossRef]
  25. Martin, A.; David, V.; Laurence, J.S.; Schwarz, P.M.; Lafer, E.M.; Hedge, A.M.; Rowe, P.S.N. Degradation of MEPE, DMP1, and release of SIBLING ASARM-Peptides (Minhibins): ASARM-peptide(s) are directly responsible for defective mineralization in HYP. Endocrinology 2008, 149, 1757–1772. [Google Scholar] [CrossRef] [Green Version]
  26. Rowe, P.S.N.; de Zoysa, P.A.; Dong, R.; Wang, H.R.; White, K.E.; Econs, M.J.; Oudet, C.L. MEPE, a new gene expressed in bone marrow and tumors causing osteomalacia. Genomics 2000, 67, 54–68. [Google Scholar] [CrossRef]
  27. Rowe, P.S.N. The wrickkened pathways of FGF23, MEPE and PHEX. Crit Rev. Oral. Biol. M 2004, 15, 264–281. [Google Scholar] [CrossRef] [Green Version]
  28. Guo, R.; Rowe, P.S.N.; Liu, S.G.; Simpson, L.G.; Xiao, Z.S.; Quarles, L.D. Inhibition of MEPE cleavage by Phex. Biochem. Bioph. Res. Co. 2002, 297, 38–45. [Google Scholar] [CrossRef]
  29. Rowe, P.S.N.; Kumagai, Y.; Gutierrez, G.; Garrett, I.R.; Blacher, R.; Rosen, D.; Cundy, J.; Navvab, S.; Chen, D.; Drezner, M.K.; et al. MEPE has the properties of an osteoblastic phosphatonin and minhibin. Bone 2004, 34, 303–319. [Google Scholar] [CrossRef] [Green Version]
  30. Atkins, G.J.; Rowe, P.S.; Lim, H.P.; Welldon, K.J.; Ormsby, R.; Wijenayaka, A.R.; Zelenchuk, L.; Evdokiou, A.; Findlay, D.M. Sclerostin Is a Locally Acting Regulator of Late-Osteoblast/Preosteocyte Differentiation and Regulates Mineralization Through a MEPE-ASARM-Dependent Mechanism. J. Bone Miner. Res. 2011, 26, 1425–1436. [Google Scholar] [CrossRef] [Green Version]
  31. Kogawa, M.; Khalid, K.A.; Wijenayaka, A.R.; Ormsby, R.T.; Evdokiou, A.; Anderson, P.H.; Findlay, D.M.; Atkins, G.J. Recombinant sclerostin antagonizes effects of ex vivo mechanical loading in trabecular bone and increases osteocyte lacunar size. Am. J. Physiol. Cell Ph. 2018, 314, C53–C61. [Google Scholar] [CrossRef] [PubMed]
  32. Krause, C.; Korchynskyi, O.; de Rooij, K.; Weidauer, S.E.; de Gorter, D.J.J.; van Bezooijen, R.L.; Hatsell, S.; Economides, A.N.; Mueller, T.D.; Lowik, C.W.G.M.; et al. Distinct Modes of Inhibition by Sclerostin on Bone Morphogenetic Protein and Wnt Signaling Pathways. J. Biol. Chem. 2010, 285, 41614–41626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Chandra, A.; Lin, T.; Young, T.; Tong, W.; Ma, X.Y.; Tseng, W.J.; Kramer, I.; Kneissel, M.; Levine, M.A.; Zhang, Y.J.; et al. Suppression of Sclerostin Alleviates Radiation-Induced Bone Loss by Protecting Bone-Forming Cells and Their Progenitors Through Distinct Mechanisms. J. Bone Miner. Res. 2017, 32, 360–372. [Google Scholar] [CrossRef]
  34. Sutherland, M.K.; Geoghegan, J.C.; Yu, C.P.; Turcott, E.; Skonier, J.E.; Winkler, D.G.; Latham, J.A. Sclerostin promotes the apoptosis of human osteoblastic cells: A novel regulation of bone formation. Bone 2004, 35, 828–835. [Google Scholar] [CrossRef] [PubMed]
  35. Karsdal, M.A.; Larsen, L.; Engsig, M.T.; Lou, H.; Ferreras, M.; Lochter, A.; Delaisse, J.M.; Foged, N.T. Matrix metalloproteinase-dependent activation of latent transforming growth factor-beta controls the conversion of osteoblasts into osteocytes by blocking osteoblast apoptosis. J. Biol. Chem. 2002, 277, 44061–44067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Kollet, O.; Dar, A.; Shivtiel, S.; Kalinkovich, A.; Lapid, K.; Sztainberg, Y.; Tesio, M.; Samstein, R.M.; Goichberg, P.; Spiegel, A.; et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat. Med. 2006, 12, 657–664. [Google Scholar] [CrossRef]
  37. Everts, V.; Delaisse, J.M.; Korper, W.; Jansen, D.C.; Tigchelaar-Gutter, W.; Saftig, P.; Beertsen, W. The bone lining cell: Its role in cleaning Howship’s lacunae and initiating bone formation. J. Bone Miner. Res. 2002, 17, 77–90. [Google Scholar] [CrossRef]
  38. Ominsky, M.S.; Niu, Q.T.; Li, C.Y.; Li, X.D.; Ke, H.Z. Tissue-Level Mechanisms Responsible for the Increase in Bone Formation and Bone Volume by Sclerostin Antibody. J. Bone Miner. Res. 2014, 29, 1424–1430. [Google Scholar] [CrossRef]
  39. Kim, S.W.; Lu, Y.H.; Williams, E.A.; Lai, F.; Lee, J.Y.; Enishi, T.; Balani, D.H.; Ominsky, M.S.; Ke, H.Z.; Kronenberg, H.M.; et al. Sclerostin Antibody Administration Converts Bone Lining Cells Into Active Osteoblasts. J. Bone Miner. Res. 2017, 32, 892–901. [Google Scholar] [CrossRef]
  40. Irie, K.; Ejiri, S.; Sakakura, Y.; Shibui, T.; Yajima, T. Matrix mineralization as a trigger for osteocyte maturation. J. Histochem. Cytochem. 2008, 56, 561–567. [Google Scholar] [CrossRef] [Green Version]
  41. Genetos, D.C.; Yellowley, C.E.; Loots, G.G. Prostaglandin E2 signals through PTGER2 to regulate sclerostin expression. PLoS ONE 2011, 6, e17772. [Google Scholar] [CrossRef] [PubMed]
  42. Winkler, D.G.; Sutherland, M.K.; Geoghegan, J.C.; Yu, C.; Hayes, T.; Skonier, J.E.; Shpektor, D.; Jonas, M.; Kovacevich, B.R.; Staehling-Hampton, K.; et al. Osteocyte control of bone formation via sclerostin, a novel BMP antagonist. Embo J. 2003, 22, 6267–6276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kogawa, M.; Wijenayaka, A.R.; Ormsby, R.T.; Thomas, G.P.; Anderson, P.H.; Bonewald, L.F.; Findlay, D.M.; Atkins, G.J. Sclerostin regulates release of bone mineral by osteocytes by induction of carbonic anhydrase 2. J. Bone Miner. Res. 2013, 28, 2436–2448. [Google Scholar] [CrossRef] [PubMed]
  44. Qing, H.; Ardeshirpour, L.; Pajevic, P.D.; Dusevich, V.; Jahn, K.; Kato, S.; Wysolmerski, J.; Bonewald, L.F. Demonstration of osteocytic perilacunar/canalicular remodeling in mice during lactation. J. Bone Miner. Res. 2012, 27, 1018–1029. [Google Scholar] [CrossRef] [Green Version]
  45. Tsourdi, E.; Jahn, K.; Rauner, M.; Busse, B.; Bonewald, L.F. Physiological and pathological osteocytic osteolysis. J. Musculoskelet Neuronal Interact. 2018, 18, 292–303. [Google Scholar]
  46. Wei, W.; Zeve, D.; Suh, J.M.; Wang, X.; Du, Y.; Zerwekh, J.E.; Dechow, P.C.; Graff, J.M.; Wan, Y. Biphasic and dosage-dependent regulation of osteoclastogenesis by beta-catenin. Mol. Cell Biol. 2011, 31, 4706–4719. [Google Scholar] [CrossRef] [Green Version]
  47. Wijenayaka, A.R.; Kogawa, M.; Lim, H.P.; Bonewald, L.F.; Findlay, D.M.; Atkins, G.J. Sclerostin stimulates osteocyte support of osteoclast activity by a RANKL-dependent pathway. PLoS ONE 2011, 6, e25900. [Google Scholar] [CrossRef] [Green Version]
  48. Weivoda, M.M.; Ruan, M.; Hachfeld, C.M.; Pederson, L.; Howe, A.; Davey, R.A.; Zajac, J.D.; Kobayashi, Y.; Williams, B.O.; Westendorf, J.J.; et al. Wnt Signaling Inhibits Osteoclast Differentiation by Activating Canonical and Noncanonical cAMP/PKA Pathways. J. Bone Miner. Res. 2016, 31, 65–75. [Google Scholar] [CrossRef] [Green Version]
  49. Holmen, S.L.; Zylstra, C.R.; Mukherjee, A.; Sigler, R.E.; Faugere, M.C.; Bouxsein, M.L.; Deng, L.F.; Clemens, T.L.; Williams, B.O. Essential role of beta-catenin in postnatal bone acquisition. J. Biol. Chem. 2005, 280, 21162–21168. [Google Scholar] [CrossRef] [Green Version]
  50. Giachelli, C.M. Ectopic calcification: Gathering hard facts about soft tissue mineralization. Am. J. Pathol. 1999, 154, 671–675. [Google Scholar] [CrossRef]
  51. Shanahan, C.M.; Crouthamel, M.H.; Kapustin, A.; Giachelli, C.M. Arterial Calcification in Chronic Kidney Disease: Key Roles for Calcium and Phosphate. Circ. Res. 2011, 109, 697–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Neven, E.; De Schutter, T.M.; De Broe, M.E.; D’Haese, P.C. Cell biological and physicochemical aspects of arterial calcification. Kidney Int. 2011, 79, 1166–1177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Neven, E.; Persy, V.; Dauwe, S.; De Schutter, T.; De Broe, M.E.; D’Haese, P.C. Chondrocyte Rather Than Osteoblast Conversion of Vascular Cells Underlies Medial Calcification in Uremic Rats. Arterioscl Throm Vas 2010, 30, 1741–1750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Barrett, H.E.; Van der Heiden, K.; Farrell, E.; Gijsen, F.J.H.; Akyildiz, A.C. Calcifications in atherosclerotic plaques and impact on plaque biomechanics. J. Biomech. 2019, 87, 1–12. [Google Scholar] [CrossRef]
  55. Goodwin, A.M.; Sullivan, K.M.; D’Amore, P.A. Cultured endothelial cells display endogenous activation of the canonical Wnt signaling pathway and express multiple ligands, receptors, and secreted modulators of Wnt signaling. Dev. Dyn. 2006, 235, 3110–3120. [Google Scholar] [CrossRef]
  56. Goodwin, A.M.; Kitajewski, J.; D’Amore, P.A. Wnt1 and Wnt5a affect endothelial proliferation and capillary length; Wnt2 does not. Growth Factors 2007, 25, 25–32. [Google Scholar] [CrossRef]
  57. Wright, M.; Aikawa, M.; Szeto, W.; Papkoff, J. Identification of a Wnt-responsive signal transduction pathway in primary endothelial cells. Biochem Biophys Res. Commun. 1999, 263, 384–388. [Google Scholar] [CrossRef]
  58. Kim, J.; Kim, J.; Kim, D.W.; Ha, Y.; Ihm, M.H.; Kim, H.; Song, K.; Lee, I. Wnt5a induces endothelial inflammation via beta-catenin-independent signaling. J. Immunol. 2010, 185, 1274–1282. [Google Scholar] [CrossRef]
  59. Vikram, A.; Kim, Y.R.; Kumar, S.; Naqvi, A.; Hoffman, T.A.; Kumar, A.; Miller, F.J., Jr.; Kim, C.S.; Irani, K. Canonical Wnt signaling induces vascular endothelial dysfunction via p66Shc-regulated reactive oxygen species. Arterioscler Thromb Vasc. Biol. 2014, 34, 2301–2309. [Google Scholar] [CrossRef] [Green Version]
  60. Wang, F.; Liu, Z.; Park, S.H.; Gwag, T.; Lu, W.; Ma, M.; Sui, Y.; Zhou, C. Myeloid beta-Catenin Deficiency Exacerbates Atherosclerosis in Low-Density Lipoprotein Receptor-Deficient Mice. Arterioscler Thromb Vasc. Biol. 2018, 38, 1468–1478. [Google Scholar] [CrossRef]
  61. Schaale, K.; Neumann, J.; Schneider, D.; Ehlers, S.; Reiling, N. Wnt signaling in macrophages: Augmenting and inhibiting mycobacteria-induced inflammatory responses. Eur. J. Cell Biol. 2011, 90, 553–559. [Google Scholar] [CrossRef] [PubMed]
  62. Borrell-Pages, M.; Romero, J.C.; Badimon, L. Cholesterol modulates LRP5 expression in the vessel wall. Atherosclerosis 2014, 235, 363–370. [Google Scholar] [CrossRef] [PubMed]
  63. Magoori, K.; Kang, M.J.; Ito, M.R.; Kakuuchi, H.; Ioka, R.X.; Kamataki, A.; Kim, D.H.; Asaba, H.; Iwasaki, S.; Takei, Y.A.; et al. Severe hypercholesterolemia, impaired fat tolerance, and advanced atherosclerosis in mice lacking both low density lipoprotein receptor-related protein 5 and apolipoprotein E. J. Biol. Chem. 2003, 278, 11331–11336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Marchand, A.; Atassi, F.; Gaaya, A.; Leprince, P.; Le Feuvre, C.; Soubrier, F.; Lompre, A.M.; Nadaud, S. The Wnt/beta-catenin pathway is activated during advanced arterial aging in humans. Aging Cell 2011, 10, 220–232. [Google Scholar] [CrossRef] [PubMed]
  65. Ezan, J.; Leroux, L.; Barandon, L.; Dufourcq, P.; Jaspard, W.; Moreau, C.; Allieres, C.; Daret, D.; Couffinhal, T.; Duplaa, C. FrzA/sFRP-1, a secreted antagonist of the Wnt-Frizzled pathway, controls vascular cell proliferation in vitro and in vivo. Cardiovasc Res. 2004, 63, 731–738. [Google Scholar] [CrossRef]
  66. Leto, G.; D’Onofrio, L.; Lucantoni, F.; Zampetti, S.; Campagna, G.; Foffi, C.; Moretti, C.; Carlone, A.; Palermo, A.; Leopizzi, M.; et al. Sclerostin is expressed in the atherosclerotic plaques of patients who undergoing carotid endarterectomy. Diabetes Metab Res. 2019, 35, e3069. [Google Scholar] [CrossRef] [Green Version]
  67. Morales-Santana, S.; Garcia-Fontana, B.; Garcia-Martin, A.; Rozas-Moreno, P.; Garcia-Salcedo, J.A.; Reyes-Garcia, R.; Munoz-Torres, M. Atherosclerotic Disease in Type 2 Diabetes Is Associated With an Increase in Sclerostin Levels. Diabetes Care 2013, 36, 1667–1674. [Google Scholar] [CrossRef] [Green Version]
  68. Krishna, S.M.; Seto, S.W.; Jose, R.J.; Li, J.; Morton, S.K.; Biros, E.; Wang, Y.; Nsengiyumva, V.; Lindeman, J.H.N.; Loots, G.G.; et al. Wnt Signaling Pathway Inhibitor Sclerostin Inhibits Angiotensin II-Induced Aortic Aneurysm and Atherosclerosis. Arterioscl Throm Vas 2017, 37, 553. [Google Scholar] [CrossRef] [Green Version]
  69. Nakamura, S.; Ishibashi-Ueda, H.; Niizuma, S.; Yoshihara, F.; Horio, T.; Kawano, Y. Coronary Calcification in Patients with Chronic Kidney Disease and Coronary Artery Disease. Clin. J. Am. Soc. Nephro 2009, 4, 1892–1900. [Google Scholar] [CrossRef]
  70. Saxena, A.; Waddell, I.C.; Friesen, R.W.; Michalski, R.T. Monckeberg medial calcific sclerosis mimicking malignant calcification pattern at mammography. J. Clin. Pathol. 2005, 58, 447–448. [Google Scholar]
  71. Castillo, B.V.; Torczynski, E.; Edward, D.P. Monckeberg’s sclerosis in temporal artery biopsy specimens. Brit. J. Ophthalmol. 1999, 83, 1091–1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Liu, J.Y.; Zhang, L.; Zhou, Y.; Zhu, D.; Wang, Q.; Hao, L.R. Aberrant activation of Wnt pathways in arteries associates with vascular calcification in chronic kidney disease. Int. Urol. Nephrol. 2016, 48, 1313–1319. [Google Scholar] [CrossRef] [PubMed]
  73. He, F.; Wang, H.; Ren, W.Y.; Ma, Y.; Liao, Y.P.; Zhu, J.H.; Cui, J.; Deng, Z.L.; Su, Y.X.; Gan, H.; et al. BMP9/COX-2 axial mediates high phosphate-induced calcification in vascular smooth muscle cells via Wnt/beta-catenin pathway. J. Cell. Biochem. 2018, 119, 2851–2863. [Google Scholar] [CrossRef] [PubMed]
  74. Jin, X.C.; Rong, S.; Yuan, W.J.; Gu, L.J.; Jia, J.S.; Wang, L.; Yu, H.L.; Zhuge, Y.F. Frontiers in Physiology High Mobility Group Box 1 Promotes Aortic Calcification in Chronic Kidney Disease via the Wnt/beta-Catenin Pathway. Front. Physiol. 2018, 9, 665. [Google Scholar] [CrossRef]
  75. Zhang, H.; Chen, J.; Shen, Z.; Gu, Y.; Xu, L.; Hu, J.; Zhang, X.; Ding, X. Indoxyl sulfate accelerates vascular smooth muscle cell calcification via microRNA-29b dependent regulation of Wnt/beta-catenin signaling. Toxicol. Lett. 2018, 284, 29–36. [Google Scholar] [CrossRef]
  76. Deng, D.; Diao, Z.; Han, X.; Liu, W. Secreted Frizzled-Related Protein 5 Attenuates High Phosphate-Induced Calcification in Vascular Smooth Muscle Cells by Inhibiting the Wnt/ss-Catenin Pathway. Calcif. Tissue Int. 2016, 99, 66–75. [Google Scholar] [CrossRef]
  77. Tian, B.Y.; Yao, L.; Sheng, Z.T.; Wan, P.Z.; Qiu, X.B.; Wang, J.; Xu, T.H. Specific knockdown of WNT8b expression protects against phosphate-induced calcification in vascular smooth muscle cells by inhibiting the Wnt-beta-catenin signaling pathway. J. Cell Physiol. 2019, 234, 3469–3477. [Google Scholar] [CrossRef]
  78. Rong, S.; Zhao, X.Z.; Jin, X.C.; Zhang, Z.; Chen, L.; Zhu, Y.X.; Yuan, W.J. Vascular Calcification in Chronic Kidney Disease is Induced by Bone Morphogenetic Protein-2 via a Mechanism Involving the Wnt/beta-Catenin Pathway. Cell Physiol. Biochem. 2014, 34, 2049–2060. [Google Scholar] [CrossRef]
  79. Freise, C.; Kretzschmar, N.; Querfeld, U. Wnt signaling contributes to vascular calcification by induction of matrix metalloproteinases. BMC Cardiovasc Disor. 2016, 16, 185. [Google Scholar] [CrossRef] [Green Version]
  80. Liao, R.X.; Wang, L.Y.; Li, J.M.; Xiong, Y.Q.; Li, Y.P.; Han, M.; Jiang, H.; Anil, M.; Su, B.H. Vascular Calcification Is Associated with Wnt-Signaling Pathway and Blood Pressure Variability in Chronic Kidney Disease Rats. Nephrol Dial. Transpl. 2019, 34. [Google Scholar] [CrossRef]
  81. Nie, B.; Zhang, S.Y.; Guan, S.M.; Zhou, S.Q.; Fang, X. Role of Wnt/beta-Catenin Pathway in the Arterial Medial Calcification and Its Effect on the OPG/RANKL System. Curr Med. Sci. 2019, 39, 28–36. [Google Scholar] [CrossRef] [PubMed]
  82. Faverman, L.; Mikhaylova, L.; Malmquist, J.; Nurminskaya, M. Extracellular transglutaminase 2 activates beta-catenin signaling in calcifying vascular smooth muscle cells. Febs Lett. 2008, 582, 1552–1557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Zeadin, M.G.; Butcher, M.K.; Shaughnessy, S.G.; Werstuck, G.H. Leptin promotes osteoblast differentiation and mineralization of primary cultures of vascular smooth muscle cells by inhibiting glycogen synthase kinase (GSK)-3 beta. Biochem. Bioph. Res. Co 2012, 425, 924–930. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, Y.Y.; Shan, J.G.; Yang, W.G.; Zheng, H.; Xue, S. High Mobility Group Box 1 (HMGB1) Mediates High-Glucose-Induced Calcification in Vascular Smooth Muscle Cells of Saphenous Veins. Inflammation 2013, 36, 1592–1604. [Google Scholar] [CrossRef]
  85. Shao, J.S.; Cheng, S.L.; Pingsterhaus, J.M.; Charlton-Kachigian, N.; Loewy, A.R.; Towler, D.A. Msx2 promotes cardiovascular calcification by activating paracrine Wnt signals. J. Clin. Investig. 2005, 115, 1210–1220. [Google Scholar] [CrossRef] [Green Version]
  86. Cai, T.; Sun, D.Q.; Duan, Y.; Wen, P.; Dai, C.S.; Yang, J.W.; He, W.C. WNT/beta-catenin signaling promotes VSMCs to osteogenic transdifferentiation and calcification through directly modulating Runx2 gene expression. Exp. Cell Res. 2016, 345, 206–217. [Google Scholar] [CrossRef] [Green Version]
  87. De Mare, A.; Maudsley, S.; Azmi, A.; Hendrickx, J.O.; Opdebeeck, B.; Neven, E.; D’Haese, P.C.; Verhulst, A. Sclerostin as Regulatory Molecule in Vascular Media Calcification and the Bone-Vascular Axis. Toxins 2019, 11, 428. [Google Scholar] [CrossRef] [Green Version]
  88. Wang, J.; Qiu, X.B.; Xu, T.H.; Sheng, Z.T.; Yao, L. Sclerostin/Receptor Related Protein 4 and Ginkgo Biloba Extract Alleviates beta-Glycerophosphate-Induced Vascular Smooth Muscle Cell Calcification By Inhibiting Wnt/beta-Catenin Pathway. Blood Purificat 2019, 47, 17–23. [Google Scholar] [CrossRef]
  89. De Oca, A.M.; Guerrero, F.; Martinez-Moreno, J.M.; Madueno, J.A.; Herencia, C.; Peralta, A.; Almaden, Y.; Lopez, I.; Aguilera-Tejero, E.; Gundlach, K.; et al. Magnesium Inhibits Wnt/beta-Catenin Activity and Reverses the Osteogenic Transformation of Vascular Smooth Muscle Cells. PLoS ONE 2014, 9, e89525. [Google Scholar] [CrossRef]
  90. Herencia, C.; Rodriguez-Ortiz, M.E.; Munoz-Castaneda, J.R.; Martinez-Moreno, J.M.; Canalejo, R.; Montes de Oca, A.; Diaz-Tocados, J.M.; Peralbo-Santaella, E.; Marin, C.; Canalejo, A.; et al. Angiotensin II prevents calcification in vascular smooth muscle cells by enhancing magnesium influx. Eur. J. Clin. Investig. 2015, 45, 1129–1144. [Google Scholar] [CrossRef]
  91. Chen, T.L.; Mao, H.J.; Chen, C.; Wu, L.; Wang, N.N.; Zhao, X.F.; Qian, J.; Xing, C.Y. The Role and Mechanism of alpha-Klotho in the Calcification of Rat Aortic Vascular Smooth Muscle Cells. Biomed. Res. Int. 2015, 194362. [Google Scholar] [CrossRef] [Green Version]
  92. Saito, Y.; Nakamura, K.; Miura, D.; Yunoki, K.; Miyoshi, T.; Yoshida, M.; Kawakita, N.; Kimura, T.; Kondo, M.; Sarashina, T.; et al. Suppression of Wnt Signaling and Osteogenic Changes in Vascular Smooth Muscle Cells by Eicosapentaenoic Acid. Nutrients 2017, 9, 858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Gao, M.; Chen, T.L.; Wu, L.; Zhao, X.F.; Mao, H.J.; Xing, C.Y. Effect of pioglitazone on the calcification of rat vascular smooth muscle cells through the downregulation of the Wnt/beta-catenin signaling pathway. Mol. Med. Rep. 2017, 16, 6208–6213. [Google Scholar] [CrossRef] [PubMed]
  94. Freise, C.; Bobb, V.; Querfeld, U. Collagen XIV and a related recombinant fragment protect human vascular smooth muscle cells from calcium-/phosphate-induced osteochondrocytic transdifferentiation. Exp. Cell Res. 2017, 358, 242–252. [Google Scholar] [CrossRef]
  95. Guo, Y.; Bao, S.M.; Guo, W.; Diao, Z.L.; Wang, L.Y.; Han, X.; Guo, W.K.; Liu, W.H. Bone marrow mesenchymal stem cell-derived exosomes alleviate high phosphorus-induced vascular smooth muscle cells calcification by modifying microRNA profiles. Funct Integr. Genom. 2019, 19, 633–643. [Google Scholar] [CrossRef]
  96. Chaturvedi, P.; Chen, N.X.; O’Neill, K.; McClintick, J.N.; Moe, S.M.; Janga, S.C. Differential miRNA Expression in Cells and Matrix Vesicles in Vascular Smooth Muscle Cells from Rats with Kidney Disease. PLoS ONE 2015, 10, e0131589. [Google Scholar] [CrossRef] [Green Version]
  97. Cheng, S.L.; Shao, J.S.; Halstead, L.R.; Distelhorst, K.; Sierra, O.; Towler, D.A. Activation of Vascular Smooth Muscle Parathyroid Hormone Receptor Inhibits Wnt/beta-Catenin Signaling and Aortic Fibrosis in Diabetic Arteriosclerosis. Circ. Res. 2010, 107, 271. [Google Scholar] [CrossRef] [Green Version]
  98. Zhang, P.; Li, Y.; Du, Y.; Li, G.; Wang, L.; Zhou, F. Resveratrol Ameliorated Vascular Calcification by Regulating Sirt-1 and Nrf2. Transpl. P 2016, 48, 3378–3386. [Google Scholar] [CrossRef]
  99. Bartoli-Leonard, F.; Wilkinson, F.L.; Langford-Smith, A.W.W.; Alexander, M.Y.; Weston, R. The Interplay of SIRT1 and Wnt Signaling in Vascular Calcification. Front. Cardiovasc Med. 2018, 5, 183. [Google Scholar] [CrossRef]
  100. Choi, S.Y.; Ryu, H.M.; Oh, E.J.; Choi, J.Y.; Cho, J.H.; Kim, C.D.; Kim, Y.L.; Park, S.H. Dipeptidyl peptidase-4 inhibitor gemigliptin protects against vascular calcification in an experimental chronic kidney disease and vascular smooth muscle cells. PLoS ONE 2017, 12, e0180393. [Google Scholar] [CrossRef] [Green Version]
  101. Shalhoub, V.; Shatzen, E.; Henley, C.; Boedigheimer, M.; McNinch, J.; Manoukian, R.; Damore, M.; Fitzpatrick, D.; Haas, K.; Twomey, B.; et al. Calcification inhibitors and Wnt signaling proteins are implicated in bovine artery smooth muscle cell calcification in the presence of phosphate and vitamin D sterols. Calcif. Tissue Int. 2006, 79, 431–442. [Google Scholar] [CrossRef] [PubMed]
  102. Martinez-Moreno, J.M.; Munoz-Castaneda, J.R.; Herencia, C.; de Oca, A.M.; Estepa, J.C.; Canalejo, R.; Rodriguez-Ortiz, M.E.; Perez-Martinez, P.; Aguilera-Tejero, E.; Canalejo, A.; et al. In vascular smooth muscle cells paricalcitol prevents phosphate-induced Wnt/beta-catenin activation. Am. J. Physiol. Renal 2012, 303, F1136–F1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Pelletier, S.; Dubourg, L.; Carlier, M.C.; Hadj-Aissa, A.; Fouque, D. The Relation between Renal Function and Serum Sclerostin in Adult Patients with CKD. Clin. J. Am. Soc. Nephro 2013, 8, 819–823. [Google Scholar] [CrossRef] [PubMed]
  104. Cejka, D.; Jager-Lansky, A.; Kieweg, H.; Weber, M.; Bieglmayer, C.; Haider, D.G.; Diarra, D.; Patsch, J.M.; Kainberger, F.; Bohle, B.; et al. Sclerostin serum levels correlate positively with bone mineral density and microarchitecture in haemodialysis patients. Nephrol Dial. Transpl. 2012, 27, 226–230. [Google Scholar] [CrossRef] [PubMed]
  105. Cejka, D.; Marculescu, R.; Kozakowski, N.; Plischke, M.; Reiter, T.; Gessl, A.; Haas, M. Renal Elimination of Sclerostin Increases With Declining Kidney Function. J. Clin. Endocr. Metab. 2014, 99, 248–255. [Google Scholar] [CrossRef]
  106. Sabbagh, Y.; Graciolli, F.G.; O’Brien, S.; Tang, W.; dos Reis, L.M.; Ryan, S.; Phillips, L.; Boulanger, J.; Song, W.P.; Bracken, C.; et al. Repression of osteocyte Wnt/beta-catenin signaling is an early event in the progression of renal osteodystrophy. J. Bone Miner. Res. 2012, 27, 1757–1772. [Google Scholar] [CrossRef]
  107. Qureshi, A.R.; Olauson, H.; Witasp, A.; Haarhaus, M.; Brandenburg, V.; Wernerson, A.; Lindholm, B.; Soderberg, M.; Wennberg, L.; Nordfors, L.; et al. Increased circulating sclerostin levels in end-stage renal disease predict biopsy-verified vascular medial calcification and coronary artery calcification. Kidney Int. 2015, 88, 1356–1364. [Google Scholar] [CrossRef] [Green Version]
  108. Bisson, S.K.; Ung, R.V.; Picard, S.; Valade, D.; Agharazii, M.; Lariviere, R.; Mac-Way, F. High calcium, phosphate and calcitriol supplementation leads to an osteocyte-like phenotype in calcified vessels and bone mineralisation defect in uremic rats. J. Bone Miner. Metab 2019, 37, 212–223. [Google Scholar] [CrossRef]
  109. Carrillo-Lopez, N.; Panizo, S.; Alonso-Montes, C.; Martinez-Arias, L.; Avello, N.; Sosa, P.; Dusso, A.S.; Cannata-Andia, J.B.; Naves-Diaz, M. High-serum phosphate and parathyroid hormone distinctly regulate bone loss and vascular calcification in experimental chronic kidney disease. Nephrol Dial. Transpl. 2019, 34, 934–941. [Google Scholar] [CrossRef]
  110. Kramann, R.; Brandenburg, V.M.; Schurgers, L.J.; Ketteler, M.; Westphal, S.; Leisten, I.; Bovi, M.; Jahnen-Dechent, W.; Knuchel, R.; Floege, J.; et al. Novel insights into osteogenesis and matrix remodelling associated with calcific uraemic arteriolopathy. Nephrol Dial. Transpl. 2013, 28, 856–868. [Google Scholar] [CrossRef] [Green Version]
  111. Brandenburg, V.M.; Kramann, R.; Rothe, H.; Kaesler, N.; Korbiel, J.; Specht, P.; Schmitz, S.; Kruger, T.; Floege, J.; Ketteler, M. Calcific uraemic arteriolopathy (calciphylaxis): Data from a large nationwide registry. Nephrol Dial. Transpl. 2017, 32, 126–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Ho, T.Y.; Chen, N.C.; Hsu, C.Y.; Huang, C.W.; Lee, P.T.; Chou, K.J.; Fang, H.C.; Chen, C.L. Evaluation of the association of Wnt signaling with coronary artery calcification in patients on dialysis with severe secondary hyperparathyroidism. BMC Nephrol. 2019, 20, 345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kuipers, A.L.; Miljkovic, I.; Carr, J.J.; Terry, J.G.; Nestlerode, C.S.; Ge, Y.R.; Bunker, C.H.; Patrick, A.L.; Zmuda, J.M. Association of circulating sclerostin with vascular calcification in Afro-Caribbean men. Atherosclerosis 2015, 239, 218–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Morena, M.; Jaussent, I.; Dupuy, A.M.; Bargnoux, A.S.; Kuster, N.; Chenine, L.; Leray-Moragues, H.; Klouche, K.; Vernhet, H.; Canaud, B.; et al. Osteoprotegerin and sclerostin in chronic kidney disease prior to dialysis: Potential partners in vascular calcifications. Nephrol. Dial. Transpl. 2015, 30, 1345–1356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Pelletier, S.; Confavreux, C.B.; Haesebaert, J.; Guebre-Egziabher, F.; Bacchetta, J.; Carlier, M.C.; Chardon, L.; Laville, M.; Chapurlat, R.; London, G.M.; et al. Serum sclerostin: The missing link in the bone-vessel cross-talk in hemodialysis patients? Osteoporos. Int. 2015, 26, 2165–2174. [Google Scholar] [CrossRef] [PubMed]
  116. Lee, Y.T.; Ng, H.Y.; Chiu, T.T.Y.; Li, L.C.; Pei, S.N.; Kuo, W.H.; Lee, C.T. Association of bone-derived biomarkers with vascular calcification in chronic hemodialysis patients. Clin. Chim. Acta 2016, 452, 38–43. [Google Scholar] [CrossRef]
  117. Lv, W.; Guan, L.N.; Zhang, Y.; Yu, S.Q.; Cao, B.F.; Ji, Y.Q. Sclerostin as a new key factor in vascular calcification in chronic kidney disease stages 3 and 4. Int. Urol. Nephrol. 2016, 48, 2043–2050. [Google Scholar] [CrossRef]
  118. Jorgensen, H.S.; Winther, S.; Dupont, L.; Bottcher, M.; Rejnmark, L.; Hauge, E.M.; Svensson, M.; Ivarsen, P. Sclerostin is not associated with cardiovascular event or fracture in kidney transplantation candidates. Clin. Nephrol. 2018, 90, 18–26. [Google Scholar] [CrossRef]
  119. Yang, C.Y.; Chang, Z.F.; Chau, Y.P.; Chen, A.; Yang, W.C.; Yang, A.H.; Lee, O.K.S. Circulating Wnt/beta-catenin signalling inhibitors and uraemic vascular calcifications. Nephrol. Dial. Transpl. 2015, 30, 1356–1363. [Google Scholar] [CrossRef]
  120. Evenepoel, P.; Goffin, E.; Meijers, B.; Kanaan, N.; Bammens, B.; Coche, E.; Claes, K.; Jadoul, M. Sclerostin Serum Levels and Vascular Calcification Progression in Prevalent Renal Transplant Recipients. J. Clin. Endocr. Metab. 2015, 100, 4669–4676. [Google Scholar] [CrossRef] [Green Version]
  121. Borawski, J.; Zoltko, J.; Labij-Reduta, B.; Koc-Zorawska, E.; Naumnik, B. Effects of Enoxaparin on Intravascular Sclerostin Release in Healthy Men. J. Cardiovasc. Pharm T 2018, 23, 344–349. [Google Scholar] [CrossRef] [PubMed]
  122. De Mare, A.; Verhulst, A.; Cavalier, E.; Delanaye, P.; Behets, G.J.; Meijers, B.; Kuypers, D.; D’Haese, P.C.; Evenepoel, P. Clinical Inference of Serum and Bone Sclerostin Levels in Patients with End-Stage Kidney Disease. J. Clin. Med. 2019, 8, 2027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Delanaye, P.; Paquot, F.; Bouquegneau, A.; Blocki, F.; Krzesinski, J.M.; Evenepoel, P.; Pottel, H.; Cavalier, E. Sclerostin and chronic kidney disease: The assay impacts what we (thought to) know. Nephrol. Dial. Transpl. 2018, 33, 1404–1410. [Google Scholar] [CrossRef] [PubMed]
  124. Piec, I.; Washbourne, C.; Tang, J.; Fisher, E.; Greeves, J.; Jackson, S.; Fraser, W.D. How Accurate is Your Sclerostin Measurement? Comparison Between Three Commercially Available Sclerostin ELISA Kits. Calcif. Tissue Int. 2016, 98, 546–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Sato, M.; Hanafusa, N.; Kawaguchi, H.; Tsuchiya, K.; Nitta, K. A Prospective Cohort Study Showing No Association Between Serum Sclerostin Level and Mortality in Maintenance Hemodialysis Patients. Kidney Blood Press R 2018, 43, 1023–1033. [Google Scholar] [CrossRef] [PubMed]
  126. Nowak, A.; Artunc, F.; Serra, A.L.; Pollock, E.; Krayenbuhl, P.A.; Muller, C.; Friedrich, B. Sclerostin Quo Vadis?—Is This a Useful Long-Term Mortality Parameter in Prevalent Hemodialysis Patients? Kidney Blood Press R 2015, 40, 266–276. [Google Scholar] [CrossRef] [PubMed]
  127. Delanaye, P.; Krzesinski, J.M.; Warling, X.; Moonen, M.; Smelten, N.; Medart, L.; Bruyere, O.; Reginster, J.Y.; Pottel, H.; Cavalier, E. Clinical and Biological Determinants of Sclerostin Plasma Concentration in Hemodialysis Patients. Nephron Clin. Pract. 2014, 128, 127–134. [Google Scholar] [CrossRef]
  128. Goncalves, F.L.C.; Elias, R.M.; dos Reis, L.M.; Graciolli, F.G.; Zampieri, F.G.; Oliveira, R.B.; Jorgetti, V.; Moyses, R.M.A. Serum sclerostin is an independent predictor of mortality in hemodialysis patients. BMC Nephrol. 2014, 15, 190. [Google Scholar] [CrossRef] [Green Version]
  129. Drechsler, C.; Evenepoel, P.; Vervloet, M.G.; Wanner, C.; Ketteler, M.; Marx, N.; Floege, J.; Dekker, F.W.; Brandenburg, V.M.; Grp, N.S. High levels of circulating sclerostin are associated with better cardiovascular survival in incident dialysis patients: Results from the NECOSAD study. Nephrol Dial. Transpl. 2015, 30, 288–293. [Google Scholar] [CrossRef] [Green Version]
  130. Jean, G.; Chazot, C.; Bresson, E.; Zaoui, E.; Cavalier, E. High Serum Sclerostin Levels Are Associated with a Better Outcome in Haemodialysis Patients. Nephron 2016, 132, 181–190. [Google Scholar] [CrossRef]
  131. Wang, X.R.; Yuan, L.A.; Zhang, J.J.; Hao, L.; Wang, D.G. Serum sclerostin values are associated with abdominal aortic calcification and predict cardiovascular events in patients with chronic kidney disease stages 3-5D. Nephrology 2017, 22, 286–292. [Google Scholar] [CrossRef] [PubMed]
  132. Lips, L.; van Zuijdewijn, C.L.M.D.; Ter Wee, P.M.; Bots, M.L.; Blankestijn, P.J.; van den Dorpel, M.A.; Fouque, D.; de Jongh, R.; Pelletier, S.; Vervloet, M.G.; et al. Serum sclerostin: Relation with mortality and impact of hemodiafiltration. Nephrol. Dial. Transpl. 2017, 32, 1217–1223. [Google Scholar] [CrossRef] [PubMed]
  133. Persy, V.; D’Haese, P. Vascular calcification and bone disease: The calcification paradox. Trends Mol. Med. 2009, 15, 405–416. [Google Scholar] [CrossRef] [PubMed]
  134. Viaene, L.; Behets, G.J.; Claes, K.; Meijers, B.; Blocki, F.; Brandenburg, V.; Evenepoel, P.; D’Haese, P.C. Sclerostin: Another bone-related protein related to all-cause mortality in haemodialysis? Nephrol Dial. Transpl. 2013, 28, 3024–3030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Rattazzi, M.; Bertacco, E.; Del Vecchio, A.; Puato, M.; Faggin, E.; Pauletto, P. Aortic valve calcification in chronic kidney disease. Nephrol. Dial. Transpl. 2013, 28, 2968–2976. [Google Scholar] [CrossRef] [Green Version]
  136. Gu, G.J.; Chen, T.; Zhou, H.M.; Sun, K.X.; Li, J. Role of Wnt/beta-catenin signaling pathway in the mechanism of calcification of aortic valve. J. Huazhong Univ. Sci. Technol. Med. Sci. 2014, 34, 33–36. [Google Scholar] [CrossRef]
  137. Brandenburg, V.M.; Kramann, R.; Koos, R.; Kruger, T.; Schurgers, L.; Muhlenbruch, G.; Hubner, S.; Gladziwa, U.; Drechsler, C.; Ketteler, M. Relationship between sclerostin and cardiovascular calcification in hemodialysis patients: A cross-sectional study. BMC Nephrol. 2013, 14, 219. [Google Scholar] [CrossRef] [Green Version]
  138. Koos, R.; Brandenburg, V.; Mahnken, A.H.; Schneider, R.; Dohmen, G.; Autschbach, R.; Marx, N.; Kramann, R. Sclerostin as a Potential Novel Biomarker for Aortic Valve. J. Heart Valve Dis 2013, 22, 317–325. [Google Scholar] [CrossRef] [Green Version]
  139. Ji, Y.Q.; Guan, L.N.; Yu, S.X.; Yin, P.Y.; Shen, X.Q.; Sun, Z.W.; Liu, J.; Lv, W.; Yu, G.P.; Ren, C. Serum sclerostin as a potential novel biomarker for heart valve calcification in patients with chronic kidney disease. Eur Rev. Med. Pharmaco 2018, 22, 8822–8829. [Google Scholar]
  140. Munroe, P.B.; Olgunturk, R.O.; Fryns, J.P.; Van Maldergem, L.; Ziereisen, F.; Yuksel, B.; Gardiner, R.M.; Chung, E. Mutations in the gene encoding the human matrix Gla protein cause Keutel syndrome. Nat. Genet. 1999, 21, 142–144. [Google Scholar] [CrossRef]
  141. Rutsch, F.; Ruf, N.; Vaingankar, S.; Toliat, M.R.; Suk, A.; Hohne, W.; Schauer, G.; Lehmann, M.; Roscioli, T.; Schnabel, D.; et al. Mutations in ENPP1 are associated with ‘idiopathic’ infantile arterial calcification. Nat. Genet. 2003, 34, 379–381. [Google Scholar] [CrossRef] [PubMed]
  142. Nitschke, Y.; Baujat, G.; Botschen, U.; Wittkampf, T.; du Moulin, M.; Stella, J.; Le Merrer, M.; Guest, G.; Lambot, K.; Tazarourte-Pinturier, M.F.; et al. Generalized Arterial Calcification of Infancy and Pseudoxanthoma Elasticum Can Be Caused by Mutations in Either ENPP1 or ABCC6. Am. J. Hum. Genet. 2012, 90, 25–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Pendleton, A.; Johnson, M.D.; Hughes, A.; Gurley, K.A.; Ho, A.M.; Doherty, M.; Dixey, J.; Gillet, P.; Loeuille, D.; McGrath, R.; et al. Mutations in ANKH cause chondrocalcinosis. Am. J. Hum. Genet. 2002, 71, 933–940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. St Hilaire, C.; Ziegler, S.G.; Markello, T.C.; Brusco, A.; Groden, C.; Gill, F.; Carlson-Donohoe, H.; Lederman, R.J.; Chen, M.Y.; Yang, D.; et al. NT5E Mutations and Arterial Calcifications. N. Engl. J. Med. 2011, 364, 432–442. [Google Scholar] [CrossRef]
  145. Rafaelsen, S.; Johansson, S.; Raeder, H.; Bjerknes, R. Long-term clinical outcome and phenotypic variability in hyperphosphatemic familial tumoral calcinosis and hyperphosphatemic hyperostosis syndrome caused by a novel GALNT3 mutation; case report and review of the literature. BMC Genet. 2014, 15. [Google Scholar] [CrossRef] [Green Version]
  146. Hsu, S.C.; Sears, R.L.; Lemos, R.R.; Quintans, B.; Huang, A.; Spiteri, E.; Nevarez, L.; Mamah, C.; Zatz, M.; Pierce, K.D.; et al. Mutations in SLC20A2 are a major cause of familial idiopathic basal ganglia calcification. Neurogenetics 2013, 14, 11–22. [Google Scholar] [CrossRef]
  147. Zarb, Y.; Weber-Stadlbauer, U.; Kirschenbaum, D.; Kindler, D.R.; Richetto, J.; Keller, D.; Rademakers, R.; Dickson, D.W.; Pasch, A.; Byzova, T.; et al. Ossified blood vessels in primary familial brain calcification elicit a neurotoxic astrocyte response. Brain 2019, 142, 885–902. [Google Scholar] [CrossRef]
  148. Bellahcene, A.; Castronovo, V. Expression of bone matrix proteins in human breast cancer: Potential roles in microcalcification formation and in the genesis of bone metastases. Bull. Cancer 1997, 84, 17–24. [Google Scholar]
  149. Grewal, R.G.; Austin, J.H. CT demonstration of calcification in carcinoma of the lung. J. Comput. Assist. Tomogr. 1994, 18, 867–871. [Google Scholar] [CrossRef]
  150. Kakkos, S.K.; Scopa, C.D.; Chalmoukis, A.K.; Karachalios, D.A.; Spiliotis, J.D.; Harkoftakis, J.G.; Karavias, D.D.; Androulakis, J.A.; Vagenakis, A.G. Relative risk of cancer in sonographically detected thyroid nodules with calcifications. J. Clin. Ultrasound 2000, 28, 347–352. [Google Scholar] [CrossRef]
  151. Klimas, R.; Bennett, B.; Gardner, W.A., Jr. Prostatic calculi: A review. Prostate 1985, 7, 91–96. [Google Scholar] [CrossRef] [PubMed]
  152. Livingston, J.H.; Stivaros, S.; Warren, D.; Crow, Y.J. Intracranial calcification in childhood: A review of aetiologies and recognizable phenotypes. Dev. Med. Child. Neurol. 2014, 56, 612–626. [Google Scholar] [CrossRef] [PubMed]
  153. Lemos, R.R.; Ferreira, J.B.; Keasey, M.P.; Oliveira, J.R. An update on primary familial brain calcification. Int Rev. Neurobiol. 2013, 110, 349–371. [Google Scholar] [CrossRef] [PubMed]
  154. Westenberger, A.; Balck, A.; Klein, C. Primary familial brain calcifications: Genetic and clinical update. Curr. Opin. Neurol. 2019, 32, 571–578. [Google Scholar] [CrossRef] [PubMed]
  155. Hassanein, A.M.; Glanz, S.M.; Kessler, H.P.; Eskin, T.A.; Liu, C. Beta-catenin is expressed aberrantly in tumors expressing shadow cells—Pilomatricoma, craniopharyngioma, and calcifying odontogenic cyst. Am. J. Clin. Pathol 2003, 120, 732–736. [Google Scholar] [CrossRef] [PubMed]
  156. Juca, C.E.B.; Colli, L.M.; Martins, C.S.; Campanini, M.L.; Paixao, B.; Juca, R.V.; Saggioro, F.P.; de Oliveira, R.S.; Moreira, A.C.; Machado, H.R.; et al. Impact of the Canonical Wnt Pathway Activation on the Pathogenesis and Prognosis of Adamantinomatous Craniopharyngiomas. Horm Metab Res. 2018, 50, 575–581. [Google Scholar] [CrossRef]
  157. Kaplon, H.; Muralidharan, M.; Schneider, Z.; Reichert, J.M. Antibodies to watch in 2020. MAbs 2020, 12, 1703531. [Google Scholar] [CrossRef] [Green Version]
  158. Brandenburg, V.M.; Verhulst, A.; Babler, A.; D’Haese, P.C.; Evenepoel, P.; Kaesler, N. Sclerostin in chronic kidney disease-mineral bone disorder think first before you block it! Nephrol. Dial. Transpl. 2019, 34, 408–414. [Google Scholar] [CrossRef]
  159. Saag, K.G.; Petersen, J.; Brandi, M.L.; Karaplis, A.C.; Lorentzon, M.; Thomas, T.; Maddox, J.; Fan, M.; Meisner, P.D.; Grauer, A. Romosozumab or Alendronate for Fracture Prevention in Women with Osteoporosis. N. Engl. J. Med. 2017, 377, 1417–1427. [Google Scholar] [CrossRef] [Green Version]
  160. Ukita, M.; Yamaguchi, T.; Ohata, N.; Tamura, M. Sclerostin Enhances Adipocyte Differentiation in 3T3-L1 Cells. J. Cell. Biochem. 2016, 117, 1419–1428. [Google Scholar] [CrossRef] [Green Version]
  161. Fairfield, H.; Falank, C.; Harris, E.; Demambro, V.; McDonald, M.; Pettitt, J.A.; Mohanty, S.T.; Croucher, P.; Kramer, I.; Kneissel, M.; et al. The skeletal cell-derived molecule sclerostin drives bone marrow adipogenesis. J. Cell Physiol. 2018, 233, 1156–1167. [Google Scholar] [CrossRef] [PubMed]
  162. Beier, E.E.; Sheu, T.J.; Resseguie, E.A.; Takahata, M.; Awad, H.A.; Cory-Slechta, D.A.; Puzas, J.E. Sclerostin activity plays a key role in the negative effect of glucocorticoid signaling on osteoblast function in mice. Bone Res. 2017, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Kim, S.P.; Frey, J.L.; Li, Z.; Kushwaha, P.; Zoch, M.L.; Tomlinson, R.E.; Da, H.; Aja, S.; Noh, H.L.; Kim, J.K.; et al. Sclerostin influences body composition by regulating catabolic and anabolic metabolism in adipocytes. Proc. Natl. Acad. Sci. USA 2017, 114, E11238–E11247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Cheng, L.J.; Zhang, L.; Yang, J.; Hao, L.R. Activation of peroxisome proliferator-activated receptor gamma inhibits vascular calcification by upregulating Klotho. Exp. Ther Med. 2017, 13, 467–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Woldt, E.; Terrand, J.; Mlih, M.; Matz, R.L.; Bruban, V.; Coudane, F.; Foppolo, S.; El Asmar, Z.; Chollet, M.E.; Ninio, E.; et al. The nuclear hormone receptor PPAR gamma counteracts vascular calcification by inhibiting Wnt5a signalling in vascular smooth muscle cells. Nat. Commun. 2012, 3. [Google Scholar] [CrossRef] [Green Version]
  166. Chouinard, L.; Felx, M.; Mellal, N.; Varela, A.; Mann, P.; Jolette, J.; Samadfam, R.; Smith, S.Y.; Locher, K.; Buntich, S.; et al. Carcinogenicity risk assessment of romosozumab: A review of scientific weight-of-evidence and findings in a rat lifetime pharmacology study. Regul. Toxicol Pharm. 2016, 81, 212–222. [Google Scholar] [CrossRef] [Green Version]
  167. Ominsky, M.S.; Boyd, S.K.; Varela, A.; Jolette, J.; Felx, M.; Doyle, N.; Mellal, N.; Smith, S.Y.; Locher, K.; Buntich, S.; et al. Romosozumab Improves Bone Mass and Strength While Maintaining Bone Quality in Ovariectomized Cynomolgus Monkeys. J. Bone Miner. Res. 2017, 32, 788–801. [Google Scholar] [CrossRef] [Green Version]
  168. Pflanz, D.; Birkhold, A.I.; Albiol, L.; Thiele, T.; Julien, C.; Seliger, A.; Thomson, E.; Kramer, I.; Kneissel, M.; Duda, G.N.; et al. Sost deficiency led to a greater cortical bone formation response to mechanical loading and altered gene expression. Sci Rep. 2017, 7. [Google Scholar] [CrossRef] [Green Version]
  169. Van Lierop, A.H.; Moester, M.J.C.; Hamdy, N.A.T.; Papapoulos, S.E. Serum Dickkopf 1 Levels in Sclerostin Deficiency. J. Clin. Endocr. Metab. 2014, 99, E252–E256. [Google Scholar] [CrossRef] [Green Version]
  170. Kang, J.H.; Keller, J.J.; Lin, H.C. Bisphosphonates reduced the risk of acute myocardial infarction: A 2-year follow-up study. Osteoporosis Int. 2013, 24, 271–277. [Google Scholar] [CrossRef]
  171. Kim, D.H.; Rogers, J.R.; Fulchino, L.A.; Kim, C.A.; Solomon, D.H.; Kim, S.C. Bisphosphonates and Risk of Cardiovascular Events: A Meta-Analysis. PLoS ONE 2015, 10. [Google Scholar] [CrossRef] [PubMed]
  172. Kranenburg, G.; Bartstra, J.W.; Weijmans, M.; de Jong, P.A.; Mali, W.P.; Verhaar, H.J.; Visseren, F.L.J.; Spiering, W. Bisphosphonates for cardiovascular risk reduction: A systematic review and meta-analysis. Atherosclerosis 2016, 252, 106–115. [Google Scholar] [CrossRef] [PubMed]
  173. EVENITY™ (romosozumab-aqqg), U.S. Prescribing Information. Available online: https://www.pi.amgen.com/~/media/amgen/repositorysites/pi-amgen-com/evenity/evenity_pi_hcp_english.ashx (accessed on 18 March 2020).
Figure 1. Overview of the actions of sclerostin in the bone. I: Inhibition of proliferation and differentiation of osteoprogenitor/pre-osteoblastic cells, as well as decreased activation of mature osteoblasts; II: decreased mineralization; III: increased apoptosis of the osteogenic cells; IV: maintenance of bone lining cells in their quiescent state; V: regulation of osteocyte maturation and osteocytic osteolysis; VI: stimulation of bone resorption.
Figure 1. Overview of the actions of sclerostin in the bone. I: Inhibition of proliferation and differentiation of osteoprogenitor/pre-osteoblastic cells, as well as decreased activation of mature osteoblasts; II: decreased mineralization; III: increased apoptosis of the osteogenic cells; IV: maintenance of bone lining cells in their quiescent state; V: regulation of osteocyte maturation and osteocytic osteolysis; VI: stimulation of bone resorption.
Ijms 21 03199 g001
Figure 2. Overview of activators and inhibitors of Wnt/β-catenin signaling pathway. BMP9: Bone-Morphogenetic Protein 9, BMP2: Bone-Morphogenetic Protein 2, Msx2: Msh homeobox 2, HMGB1: High Mobility Group Box 1, sFRP5: secreted Frizzled-Related Protein 5, PPARγ: Peroxisome Proliferator-Activated Receptor γ, PTH1R: Parathyroid Hormone 1 Receptor, SIRT1: Sirtuin1, MMP: Matrix Metalloproteinase, Runx2: Runt-related transcription factor 2, RANKL: Receptor Activator of Nuclear Factor Kappa-Β Ligand.
Figure 2. Overview of activators and inhibitors of Wnt/β-catenin signaling pathway. BMP9: Bone-Morphogenetic Protein 9, BMP2: Bone-Morphogenetic Protein 2, Msx2: Msh homeobox 2, HMGB1: High Mobility Group Box 1, sFRP5: secreted Frizzled-Related Protein 5, PPARγ: Peroxisome Proliferator-Activated Receptor γ, PTH1R: Parathyroid Hormone 1 Receptor, SIRT1: Sirtuin1, MMP: Matrix Metalloproteinase, Runx2: Runt-related transcription factor 2, RANKL: Receptor Activator of Nuclear Factor Kappa-Β Ligand.
Ijms 21 03199 g002
Figure 3. Hypothesized contribution of sclerostin in the calcification paradox. In a calcified artery, sclerostin is produced by transdifferentiated vascular smooth muscle cells [87,108]. It is hypothesized that this locally produced sclerostin can spill-over into the circulation, thereby contributing to increased serum sclerostin levels [87,134]. Via the circulation, this increased amount of sclerostin reaches the bone compartment, where it might be involved in the inhibition of bone formation/mineralization/turnover [87].
Figure 3. Hypothesized contribution of sclerostin in the calcification paradox. In a calcified artery, sclerostin is produced by transdifferentiated vascular smooth muscle cells [87,108]. It is hypothesized that this locally produced sclerostin can spill-over into the circulation, thereby contributing to increased serum sclerostin levels [87,134]. Via the circulation, this increased amount of sclerostin reaches the bone compartment, where it might be involved in the inhibition of bone formation/mineralization/turnover [87].
Ijms 21 03199 g003

Share and Cite

MDPI and ACS Style

De Maré, A.; D’Haese, P.C.; Verhulst, A. The Role of Sclerostin in Bone and Ectopic Calcification. Int. J. Mol. Sci. 2020, 21, 3199. https://doi.org/10.3390/ijms21093199

AMA Style

De Maré A, D’Haese PC, Verhulst A. The Role of Sclerostin in Bone and Ectopic Calcification. International Journal of Molecular Sciences. 2020; 21(9):3199. https://doi.org/10.3390/ijms21093199

Chicago/Turabian Style

De Maré, Annelies, Patrick C. D’Haese, and Anja Verhulst. 2020. "The Role of Sclerostin in Bone and Ectopic Calcification" International Journal of Molecular Sciences 21, no. 9: 3199. https://doi.org/10.3390/ijms21093199

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop