Novel insights on the effect of sclerostin on bone and other organs

in Journal of Endocrinology
Authors:
Timothy J Dreyer Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK

Search for other papers by Timothy J Dreyer in
Current site
Google Scholar
PubMed
Close
,
Jacob AC Keen Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK

Search for other papers by Jacob AC Keen in
Current site
Google Scholar
PubMed
Close
,
Leah M Wells Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK

Search for other papers by Leah M Wells in
Current site
Google Scholar
PubMed
Close
, and
Scott J Roberts Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, UK

Search for other papers by Scott J Roberts in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0001-7460-7292

Correspondence should be addressed to S J Roberts: sjroberts@rvc.ac.uk
Free access

Sign up for journal news

As a key regulator of bone homeostasis, sclerostin has garnered a lot of interest over the last two decades. Although sclerostin is primarily expressed by osteocytes and is well known for its role in bone formation and remodelling, it is also expressed by a number of other cells and potentially plays a role in other organs. Herein, we aim to bring together recent sclerostin research and discuss the effect of sclerostin on bone, cartilage, muscle, liver, kidney and the cardiovascular and immune systems. Particular focus is placed on its role in diseases, such as osteoporosis and myeloma bone disease, and the novel development of sclerostin as a therapeutic target. Anti-sclerostin antibodies have recently been approved for the treatment of osteoporosis. However, a cardiovascular signal was observed, prompting extensive research into the role of sclerostin in vascular and bone tissue crosstalk. The study of sclerostin expression in chronic kidney disease was followed by the investigation of its role in liver–lipid–bone interactions, and the recent discovery of sclerostin as a myokine prompted new research into sclerostin within the bone–muscle relationship. Potentially, the effects of sclerostin reach beyond that of bone alone. We further summarise recent developments in the use of sclerostin as a potential therapeutic for osteoarthritis, osteosarcoma and sclerosteosis. Overall, these new treatments and discoveries illustrate progress within the field, however, also highlight remaining gaps in our knowledge.

Abstract

As a key regulator of bone homeostasis, sclerostin has garnered a lot of interest over the last two decades. Although sclerostin is primarily expressed by osteocytes and is well known for its role in bone formation and remodelling, it is also expressed by a number of other cells and potentially plays a role in other organs. Herein, we aim to bring together recent sclerostin research and discuss the effect of sclerostin on bone, cartilage, muscle, liver, kidney and the cardiovascular and immune systems. Particular focus is placed on its role in diseases, such as osteoporosis and myeloma bone disease, and the novel development of sclerostin as a therapeutic target. Anti-sclerostin antibodies have recently been approved for the treatment of osteoporosis. However, a cardiovascular signal was observed, prompting extensive research into the role of sclerostin in vascular and bone tissue crosstalk. The study of sclerostin expression in chronic kidney disease was followed by the investigation of its role in liver–lipid–bone interactions, and the recent discovery of sclerostin as a myokine prompted new research into sclerostin within the bone–muscle relationship. Potentially, the effects of sclerostin reach beyond that of bone alone. We further summarise recent developments in the use of sclerostin as a potential therapeutic for osteoarthritis, osteosarcoma and sclerosteosis. Overall, these new treatments and discoveries illustrate progress within the field, however, also highlight remaining gaps in our knowledge.

Introduction

Sclerostin is a 22 kDa secreted glycoprotein encoded by the SOST gene. It is primarily expressed by osteocytes and plays a major role in bone homeostasis, affecting bone formation and bone remodelling through its role as a negative regulator of the Wnt/β-catenin signalling pathway (reviewed byHoldsworth et al. 2019). Sclerostin achieves this through inhibition of wingless-related integration site (Wnt)–ligand interaction with low-density lipoprotein receptor protein 4/5/6 (LRP4/5/6) Wnt co-receptors (Leupin et al. 2011, Holdsworth et al. 2012). Changes in sclerostin expression mediates its function as a negative regulator of bone formation: SOST overexpression results in decreased bone formation, mass and strength, whereas disruption of sclerostin function or expression results in high bone mass conditions (Balemans et al. 2001, Van Wesenbeeck et al. 2003, Winkler et al. 2003, Loots et al. 2005, Niziolek et al. 2015, Zhang et al. 2016, Kim et al. 2017a ). Thus, it is crucial that we continue to investigate this fascinating protein to better understand its biology and to provide future therapeutic options for disease modification.

The current review examines recent literature and expands on the earlierHoldsworth et al. (2019) review, which examined the structure and biological function of sclerostin as well as its role in disease (Holdsworth et al. 2019). We explore recent progress made in our under­standing of both sclerostin biology and its mechanism of action in the skeleton and beyond (Fig. 1). We also examine recent evidence for its potential as a therapeutic and highlight areas where further research could expand our current understanding of this protein.

Figure 1
Figure 1

Current knowledge on the role of sclerostin in the body. Primary research article searches of sclerostin and tissue type published between 2001 and 2022 were carried out on PubMed with review articles excluded. The number of hits were converted into log base 2 to generate a scale, with hits for each tissue highlighted on the scale. Arrows in the tissue detail boxes indicate direction of altered sclerostin levels in disease with colour of box reflecting the volume of data published based on the scale generated. The number of studies per tissue excluding review articles: bone (2312), kidney (282), cardiovascular (213), muscle (115), cartilage (88), immune system (61) and liver (44). Created with https://www.biorender.com/.

Citation: Journal of Endocrinology 257, 2; 10.1530/JOE-22-0209

The Wnt/β-catenin signalling pathway

Wnt inhibitors such as sclerostin and Dickkopf-related protein 1 (DKK1) elicit their biological effects through interaction with the Wnt/β-catenin signalling pathway, which has been reviewed extensively and will not be discussed in depth herein (Ke et al. 2012, Nusse & Clevers 2017). Briefly, canonical Wnt signalling occurs via an autocrine or paracrine fashion presenting in either an on or off state (Fig. 2). The signalling cascade is activated when extracellular Wnt interacts with LRP5/6 and frizzled family (FZD) cell surface receptors, forming a ternary complex. This results in phosphorylation of the LRP5/6 cytoplasmic domain and subsequent translocation via recruitment of the destruction complex consisting of axin, dishevelled (DVL), adenomatous polyposis coli (APC), glycogen synthase kinase 3β (GSK3β) and casein kinase 1 (CK1). Localisation to the cellular membrane disrupts the destruction complex activity, allowing non-phosphorylated β-catenin to accumulate in the cytoplasm. Here it is translocated to the nucleus to initiate transcription of Wnt target genes through interaction with TCF/LEF transcription factors (Ke et al. 2012, Nusse & Clevers 2017). The Wnt signalling pathway is switched to its off state when Wnt antagonists such as sclerostin and DKK1 interact with LRP5/6 surface receptors, preventing Wnt LRP5/6 interaction and subsequent formation of the Wnt/FZD/LRP5/6 ternary complex (Semënov et al. 2001, Ai et al. 2005, Balemans et al. 2008, Choi et al. 2009, Holdsworth et al. 2012, Ke et al. 2012, Bullock et al. 2019). The destruction complex remains active and ubiquitinates cytoplasmic phosphorylated β-catenin for degradation by the proteasome. β-catenin does not translocate to the nucleus, and Wnt target gene transcription is repressed due to the association of Groucho instead of β-catenin with TCF/LEF (Nusse & Clevers 2017).

Figure 2
Figure 2

Wnt/β-catenin signalling pathway. During the ‘on’ state of the pathway, Wnt binds to LRP5/6, resulting in localisation of the destruction complex to the membrane and disruption of destruction complex activity. Non-phosphorylated β-catenin accumulates in the cytoplasm and is translocated to the nucleus where it interacts with TCF/LEF to initiate transcription of Wnt target genes. During the ‘off’ state, sclerostin (Scl) binds to LRP4/5/6 and DKK1 interacts with LRP5/6 and Kremen, preventing Wnt from interacting with the Wnt receptors. Phosphorylated β-catenin is ubiquitinated by the destruction complex for degradation by the proteasome, resulting in repression of Wnt target gene expression. Created with https://www.biorender.com/.

Citation: Journal of Endocrinology 257, 2; 10.1530/JOE-22-0209

Sclerostin and bone

Activation of the Wnt/β-catenin signalling pathway promotes mesenchymal cell differentiation into preosteoblasts and subsequent osteoblasts, which lay down an organic matrix that is mineralised to form bone (reviewed byHoldsworth et al. 2019). Osteoblasts either remain on the bone surface as quiescent bone lining cells, undergo apoptosis or become embedded in the bone matrix to eventually become osteocytes, the primary source of sclerostin in the adult skeleton (Poole et al. 2005, Capulli et al. 2014). In addition to negatively regulating the above processes, sclerostin contributes to maintaining the bone lining cells in a quiescent state and inhibits differentiation of late osteoblasts into osteocytes via inhibition of canonical Wnt signalling (Atkins et al. 2011, Kim et al. 2017b , Hong et al. 2022). Sclerostin also plays an important role in the regulation of bone marrow adiposity and bone marrow adipose tissue (reviewed by Holdsworth et al. 2019). Furthermore, sclerostin indirectly affects bone resorption by upregulating osteocyte-expressed receptor activator of nuclear factor-κB ligand (RANKL), an essential cytokine for osteoclast development and activity, and decreasing expression of osteoprotegerin (OPG), a decoy receptor for RANKL, in mature osteoblasts and osteocytes, thereby increasing osteoclastogenesis (Simonet et al. 1997, Glass et al. 2005, Nakashima et al. 2011, Wijenayaka et al. 2011). Indeed, administration of anti-sclerostin antibodies to oestrogen-deficient osteocytes decreases osteoclastogenesis and resorption, highlighting the importance of sclerostin in the upregulated pro-osteoclastogenic signalling between osteocytes and osteoclasts in the absence of oestrogen (Allison et al. 2020). Moreover, the presence of osteoclasts in trabecular bone reduces sclerostin expression in osteocytes, suggesting that osteoclast-mediated reduction in sclerostin facilitates trabecular bone formation (Koide et al. 2020). Overall, these results demonstrate the importance of sclerostin in modulating the RANKL/OPG ratio, a major determinant of bone mass and strength (reviewed byHoldsworth et al. 2019).

Osteoporosis

Postmenopausal (type I) osteoporosis is the most common form of the disease and is characterised by increased bone fragility and susceptibility to fractures. These symptoms result from low bone mass and reduced bone structural integrity caused by aging and menopause-related decrease in oestrogen levels, which has been shown to increase sclerostin expression (Dobbs et al. 1999). Despite this link, studies have shown serum and bone sclerostin levels to be positively correlated with bone mineral density (BMD) in osteoporosis, which may be explained by the decreased osteocyte number in low BMD disorders such as osteoporosis (Ueland et al. 2019, Gorter et al. 2022). Osteoporosis is treated with either antiresorptive or bone-anabolic drugs. The major antiresorptives are bisphosphonates and denosumab, a humanised monoclonal anti-RANKL antibody (Storm et al. 1990, Pols et al. 1999, McClung et al. 2006, Brown et al. 2009, Baron et al. 2011). The anabolic agents teriparatide (parathyroid hormone (PTH) 1-34) and abaloparatide (parathyroid hormone-related protein (PTHrP) 1-34) are used in patients with more severe and established osteoporosis (Yu et al. 2014, Leder et al. 2015, Eastell et al. 2019). In 2019, an additional anabolic agent, romosozumab (EVENITY™), was approved by the United States Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for treating postmenopausal women at a high risk of osteoporotic fractures. Romosozumab, a humanised monoclonal anti-sclerostin antibody, blocks sclerostin-mediated Wnt inhibition, resulting in increased bone formation whilst inhibiting bone resorption. Phase III clinical trials have shown therapeutic effectiveness in reducing fractures in elderly osteoporotic patients (Cosman et al. 2016, Saag et al. 2017, Lewiecki et al. 2018). However, there are concerns regarding the effect of romosozumab on the cardiovascular system (discussed in more detail later in this review). Alternative anti-sclerostin antibodies are also in various stages of development including setrusumab, a sclerostin neutralising human IgG2λ monoclonal antibody which is being investigated for the treatment of moderate osteogenesis imperfecta and hypophosphatasia (Glorieux et al. 2017, Seefried et al. 2017). Additionally, blosozumab, a recombinant humanised antibody, has shown promising results as a potential treatment for postmenopausal osteoporosis and is currently in phase I clinical trials in China (Recker et al. 2015).

Idiopathic scoliosis

Idiopathic scoliosis (IS) is a low bone mass disorder often characterised by spinal deformities (Cheng et al. 2007). Zhang et al. reported lower SOST gene expression and serum sclerostin levels that were negatively correlated with plasma miRNA-145 in adolescent IS (AIS), the most common type of scoliosis (Zhang et al. 2018). Moreover, osteocyte secretion of sclerostin was reduced in AIS patients due to reduced osteocyte numbers as well as augmented osteocyte function caused by aberrant miRNA-145/β-catenin expression. Overactive canonical Wnt signalling in IS inhibits osteoblast differentiation to osteocytes and negatively affects matrix mineralisation in AIS instead of yielding the expected high bone mass phenotype (Rodda & McMahon 2006, Regard et al. 2011). This defect in mineralisation results in inferior bone mechanical properties which might increase the susceptibility of bone to asymmetrical forces, leading to spinal column deformities (Vasiliadis et al. 2021). Vasiliadis et al. hypothesised that stimulating osteocyte sclerostin secretion and restoring normal function of the Wnt/β-catenin signalling pathway during growth could, in theory, increase bone strength and prevent deterioration of the scoliotic deformity. However, Zhang et al. noted that further study of osteoclastogenesis and osteoclast resorption activity is required to give a more comprehensive picture of bone remodelling in AIS (Zhang et al. 2018). Further investigation of miRNA-145 effects on osteoblast and osteocyte development and function may provide clarity on AIS-related changes in SOST expression.

Myeloma bone disease

Myeloma bone disease (MBD) is characterised by a plasma cell malignancy that forms in the bone marrow, often leading to severe bone destruction, pathological fractures, osteolytic bone lesions and debilitating bone pain (Delgado-Calle et al. 2014, Paton‐Hough et al. 2019). The condition is caused by imbalance and uncoupling of the bone-remodelling process, whereby sclerostin is suggested to contribute to increased osteoclast-mediated bone resorption and decreased osteoblast-mediated bone formation (Paton‐Hough et al. 2019). Indeed, MBD patients have increased serum sclerostin, correlating with disease stage and degree of bone destruction (Eda et al. 2016). The source of this sclerostin is contentious, with multiple studies reporting that multiple myeloma (MM) cells secrete sclerostin with other osteoclast activating factors and osteoblast inhibitory factors, including DKK1 in the bone marrow micro-environment (Tian et al. 2003, Brunetti et al. 2011, Colucci et al. 2011, Habibi et al. 2013, Eda et al. 2016). Conversely, McDonald et al. reported that SOST was not expressed in MM cells isolated from myeloma patients or in numerous myeloma cell lines (McDonald et al. 2017). However, MM cancer cells have been shown to alter osteocyte (the primary source of sclerostin) viability and gene expression (Atkinson & Delgado‐Calle 2019). Liu et al. showed that sclerostin is upregulated by histone acetyltransferase major histocompatibility complex (MHC) class II transactivator (CIITA) expressed in osteocytes, resulting in decreased osteoblastogenesis and increased osteoclastogenesis (Liu et al. 2022). Furthermore, MM cells increase osteocyte death through upregulation of activated Notch signalling-mediated apoptosis and autophagy, which triggers osteoclast formation and activity (Giuliani et al. 2012, Delgado-Calle et al. 2016, Toscani et al. 2018). Notably, Mabille et al. showed that DKK1 and sclerostin are increased 4 months prior to relapse from treatment (Mabille et al. 2018).

There is a positive correlation between circulating sclerostin and osteolytic fractures, disease stage and bone remodelling markers in MBD patients, with mounting evidence for the benefit of bone anabolic agents in treatment of MBD (Gau et al. 2022). Mice injected with myeloma cells showed significant bone loss (Lawson et al. 2015). Interestingly, Sost deletion or treatment with anti-sclerostin antibodies increased osteoblastogenesis and bone formation rate. Additionally, anti-sclerostin antibodies prevented myeloma-induced bone loss, increased fracture resistance and decreased osteolytic bone lesions without interfering with MM chemotherapy (Delgado-Calle et al. 2017). Co-treatment with the antiresorptive agent zoledronic acid (Zol) increased bone mass and fracture resistance when compared with Zol alone in preclinical models of myeloma and may prevent the onset of MBD whilst increasing resistance to fractures (McDonald et al. 2017). Furthermore, combined inhibition of sclerostin and DKK1 (DKK1 levels are elevated in MM cells and rodents treated with anti-sclerostin antibodies) with a bi-specific antibody or a combination of anti-sclerostin and anti-DKK1 antibodies has a synergistic effect on increased bone formation and bone strength in mice and might be a potential therapeutic strategy for treating MBD (Stolina et al. 2014, Nioi et al. 2015, Florio et al. 2016, Taylor et al. 2016, Holdsworth et al. 2018, Witcher et al. 2018).

Sclerosteosis

In contrast to osteoporosis and IS, sclerosteosis is a high bone mass condition. It is autosomal recessive and is caused by loss of function mutations in SOST and LRP4, the latter causing sclerosteosis 2 (Beighton 1988, Balemans et al. 2001, Balemans et al. 2002, Fijalkowski et al. 2016). This condition is associated with increased bone formation and has been studied intensively in Sost –/− mice, a mouse model of sclerosteosis (Li et al. 2008). It has recently been shown that due to lack of sclerostin-mediated upregulation of RANKL expression, remodelling-based bone formation likely accounts for two-thirds of bone formed in 12-week-old Sost-deficient mice (Koide et al. 2022). Importantly, sclerosteosis has no pharmacological treatment with symptoms managed through surgery. However, efforts have been made to develop potential new treatments through protein replacement therapy (Dreyer et al. 2021).

Sclerostin and cartilage

Chondrocytes produce and maintain the cartilage extracellular matrix (ECM) present on the articular surface for healthy joint function (reviewed byAkkiraju & Nohe 2015). These stable, mature chondrocytes are derived from mesenchymal cells that differentiate down the chondrogenic lineage. Differentiation is halted once the mature chondrocyte stage is reached and cells remain in a steady state, maintaining cartilage homeostasis. In contrast, during endochondral ossification (observed during long bone growth and fracture repair), mature chondrocytes undergo hypertrophic differentiation leading to the catabolism of ECM components. This degeneration of the cartilage ECM includes mineral deposition and paves the way for subsequent bone formation (Goldring 2012). Sclerostin has been shown to be expressed in chondrocytes in vitro, facilitating early chondrogenic differentiation through suppression of Wnt/β-catenin signalling (Yamaguchi et al. 2018). This expression is lost during hypertrophic differentiation resulting in high levels of Wnt/β-catenin signalling (Ma et al. 2013, Yamaguchi et al. 2018). Interestingly, Pinch2 knockout mice have been shown to express high levels of sclerostin in their hypertrophic zone chondrocytes and phenotypically display low bone mass and shortened limbs, providing further evidence that sclerostin may be a negative regulator of endochondral ossification (Lei et al. 2020).

Osteoarthritis

Osteoarthritis (OA) is a degenerative joint disease characterised by progressive loss of articular cartilage, in addition to subchondral bone exposure and remodelling, synovial inflammation and osteophyte formation. During OA development, mature chondrocytes continue to differentiate into a hypertrophic and catabolic state which leads to degeneration of the cartilage matrix (Goldring 2012). Sclerostin expression is elevated in articular chondrocytes following surgically induced OA and is reduced in subchondral bone in association with bone sclerosis, suggesting that sclerostin might play a role in preventing articular cartilage degradation (Chan et al. 2011). Additionally, an increase in miRNA-218-5p, a SOST inhibitor, was reported to be upregulated in moderate to severe human OA and has been suggested as a potential therapeutic target (Lu et al. 2017). During OA development sclerostin expression is reduced in human subchondral bone from OA subjects undergoing total knee arthroplasty (Wu et al. 2016). This has been replicated in vivo with Sost-deficient mice displaying increased subchondral bone sclerosis and subsequent OA development (Li et al. 2019a ). It is suggested that OA osteoclasts secrete higher levels of the sclerostin-negative regulator, leukaemia inhibitory factor (LIF), resulting in increased Wnt/β-catenin signalling. This leads to subsequent abnormal bone remodelling, further exacerbating cartilage destruction. Interestingly, inhibition of osteoclasts with alendronate attenuates LIF expression and cartilage degeneration (Zhao et al. 2022). These data suggest that sclerostin depletion may play a role in OA development and is therefore a plausible therapeutic target for targeting early to moderate OA as a means to prevent disease progression.

Sclerostin and the cardiovascular system

Increasing data suggests a role for Wnt signalling in the pathophysiology of vascular diseases and ageing (Catalano et al. 2020). Wnt signalling also interacts with endothelial dysfunction, affecting both the proliferation and migration of vascular smooth muscle cells (VSMCs) and intimal thickening.

Atherosclerosis

Atherosclerosis is a chronic inflammatory disease of the arterial wall (Libby et al. 2019). Research suggests that sclerostin is linked to subclinical atherosclerosis and is inversely associated with carotid intima-media thickness (CIMT) in postmenopausal woman with type II diabetes mellitus (Morales-Santana et al. 2013, Gaudio et al. 2014). Leto et al. reported SOST expression at the vascular level in humans. Sclerostin production was identified in atherosclerotic plaques of patients that had carotid endarterectomy, with significantly greater quantities in the media compared to intima. Vascular smooth muscle cells (VSMCs) also displayed higher sclerostin levels than infiltrating macrophages (Leto et al. 2019). Excess sclerostin has been shown to be protective against the progression of both atherosclerosis and inflammation in the Apolipoprotein E (ApoE) mouse model (Krishna et al. 2017). Decreased sclerostin may therefore be related to higher susceptibility to atheroprogression. Turk et al. further evaluated the relationship between sclerostin and atheroprogression with nonclinical toxicology and safety packages and found no significant correlation between sclerostin inhibition and atheroprogression in two ApoE models (Turk et al. 2020). Similarly, Holdsworth et al. observed little or no sclerostin in human plaques. Sclerostin intensity was decreased compared to normal aorta and was found in deeper areas of the plaque and aorta wall, but not in regions known to be relevant to plaque stability, such as the fibrous cap and the endothelium. These data indicate little association of sclerostin involvement with the stability of an atherosclerotic plaque (Holdsworth et al. 2021).

Vascular calcification

Sclerostin has been discovered in the aorta of patients undergoing aortic valve replacement and found to be increased in both calcified vascular plaques and calcifying VSMCs (Didangelos et al. 2011, Zhu et al. 2011, Koos et al. 2013). Sclerostin serum levels were positively correlated with the presence of thoracic aorta calcification and positive SOST expression in the vascular system (Li et al. 2019b ). Evidence also exists that sclerostin could differentially affect vascular calcification (VC) in distinct vascular beds. Sclerostin was linked to an increased risk (1.61×) of coronary artery calcification (CAC) whilst there was no link to aortic artery calcification (AAC) in recent work performed by Kuipers et al. (2015). However, calcified aorta in rats with chronic kidney disease (CKD) secreted increased amounts of sclerostin and displayed impaired bone metabolism (Mace et al. 2021). Further in vitro analysis with calcified aorta rings co-incubated with UMR-106 osteoblast‐like cells showed detrimental effects of the calcified aorta on bone mineralisation, suggesting crosstalk between vascular and bone tissue (Mace et al. 2022). De Maré et al. showed that SOST−/− mice with adenine diet-induced CKD had significantly higher calcium content in their aorta compared to wild type (De Maré et al. 2022). DBA/2J mice on a warfarin diet were treated with an anti-sclerostin antibody and displayed significantly more calcification in both the aorta and renal arteries, suggesting that sclerostin offers a protective role during the development of vascular calcifications (De Maré et al. 2022). The bone–vascular axis should thus be a key consideration when developing new therapeutics for vascular calcification and augmented bone metabolism (De Maré et al. 2019a ).

Romosozumab and cardiovascular events

Romosozumab has been compared to alendronate, an oral bisphosphonate that effectively suppresses osteoclast-mediated bone resorption and lowers fracture risk in postmenopausal women with osteoporosis (Schenk et al. 1986, Liberman et al. 1995, Chavassieux et al. 1997, Saag et al. 2017). Moreover, bisphosphonates have previously been linked to cardioprotection; however, a recent review by Fuggle et al. suggests that this might not be the case (Kim et al. 2015, Kranenburg et al. 2016, Fuggle et al. 2020).

Data collected from BRIDGE (placebo-controlled double-blind study evaluating the efficacy and safety of romosozumab in treating men with osteoporosis) and ARCH (active-controlled fracture study in postmenopausal women with osteoporosis at high risk) phase III randomised control trials (RCTs) using romosozumab showed a disproportionate occurrence in serious cardiovascular events (CVEs) (Saag et al. 2017, Lewiecki et al. 2018). This caused regulatory bodies to issue warnings on the product labels for the increased risk of heart attack, stroke and cardiovascular deaths. As such, romosozumab is not indicated for patients who have suffered such an event in the past year.

The increased adverse CVEs observed when comparing romosozumab to alendronate (2.5% vs 1.9%) during the ARCH phase III RCT could not be explained by baseline cardiovascular risk or concurrent use of cardiovascular medication (Saag et al. 2017). The FRAME (fracture study in post-menopausal women with osteoporosis) study did not produce similar results; however, this trial did enrol a study population with different criteria and was placebo controlled (Cosman et al. 2016). A meta-analysis by Lv et al. showed that romosozumab treatment was not associated with an increased risk of a three-point major adverse cardiovascular event composite (cardiovascular death or death, myocardial infarction and stroke), composite cardiovascular outcomes (including stroke, atrial fibrillation, heart failure and coronary heart disease) or any specific cardiovascular outcomes in patients with osteoporosis (Lv et al. 2020). However, there was a significant increase in the risk of four-point major adverse cardiovascular event composite (cardiovascular death, myocardial infarction, stroke and heart failure) with romosozumab treatment. Results from another meta-analyses of both published and unpublished (FRAME data made available to the U.S. FDA Drugs Advisory Committee) cardiovascular outcome trial data from romosozumab suggested that SOST genetic variants were also at a higher risk of myocardial infarction and/or coronary revascularisation and major adverse cardiovascular events, and concluded that sclerostin inhibition could elevate the risk of cardiovascular disease (CVD), necessitating a review of the cardiovascular safety of anti-sclerostin therapies (Bovijn et al. 2020).

However, the meta-analyses received criticism from Holm et al. who found fault with both the analysis and interpretation of the results (Holm et al. 2021). This study indicated that the analysis used a non-suitable significance threshold and use of the suggested P-value would require significant backing by data, which was not provided. The meta-analysis of all the RCTs also showed no association with cardiovascular incidences. It was also noted that coronary artery disease had been examined extensively within genome-wide association studies (GWAS), yet no SOST variants were seen (Buniello et al. 2019). Furthermore, there was disagreement with reporting the P-values of the combined markers when these are not independent, thus exaggerating the significance (Holm et al. 2021). Holm et al. noted that any potential relationship between the SOST BMD variant and CV risk has not been established as a driver by Bovijn et al. There are other SOST variants found in these vascular regions, thus making it impossible to undertake colocalisation analyses with the CVD phenotype (Holm et al. 2021). Bovijn et al. rebutted the comments, cautioning that the data from the RCTs of romosozumab did not show any relationship determined by their choice of P-value (Buniello et al. 2019). Additionally, the scale of a GWAS variant is not a correct illustration of the magnitude of biological effect seen when augmenting the protein produced by the same gene with a therapeutic agent (Bovijn et al. 2021). Regardless, despite contradictory data or the lack of evidence that inhibiting sclerostin has a detrimental effect upon the cardiovascular system, it is recommended that romosozumab should be carefully prescribed, weighing up the cardiovascular risk to the patient (Langdahl et al. 2021).

Work conducted by Yu et al. examined the role of sclerostin in cardiovascular protection as well as bone formation by targeting different loops of the sclerostin structure. In vitro and in vivo studies demonstrated that loop 3 deficiency, either by pharmacologically targeting loop 3 with Apc001PE or by genetic truncation, maintained protective features on the cardiovascular system. Interestingly, both loop 2 and loop 3 targeting in MC3T3-E1 cells diminished the inhibition of bone formation by sclerostin, which was mirrored in vivo in mice. Of note, the authors did not provide comparative data to sclerostin neutralisation through the use of an approved therapeutic antibody. As such, the decrease in cardiovascular risk could not be entirely quantified. Nevertheless, these data could inform on the creation of novel sclerostin inhibitors for future bone therapeutics coupled with decreased cardiovascular risk (Yu et al. 2022).

Other cardiovascular events

The function of sclerostin in aortic aneurysms was examined in vitro as well as in mouse models and human specimens (Krishna et al. 2017). This study revealed that sclerostin expression in the aorta was downregulated in human aortic aneurysms, possibly due to epigenetic silencing. Interestingly, sclerostin prevented angiotensin II-induced aortic aneurysms in the thoracic and abdominal aorta in SOST transgenic ApoE −/− mice, suggesting a potential therapeutic strategy via upregulation of sclerostin (Krishna et al. 2017). Arterial wall stiffness is a hallmark of arterial ageing, and interestingly sclerostin has been identified as an independent marker of arterial stiffness in healthy adults. However, DKK1 did not show a similar correlation (Gaudio et al. 2017). These two Wnt signalling inhibitors have very similar roles in vascular cells yet show a significant difference in behaviour in this process, which could account for their differing expression throughout the body (Gaudio et al. 2014, Desjardins et al. 2014, Hsu et al. 2016).

Interestingly, Javaheri et al. found that bone loss in a mouse model of glucocorticoid excess was not rescued by Sost haploinsufficiency (Javaheri et al. 2019), contradicting previous studies which reported that genetic deletion of Sost and anti-sclerostin antibody treatment rescued glucocorticoid-induced low bone mass (Marenzana et al. 2011, Sato et al. 2016, Yao et al. 2016, Javaheri et al. 2019). However, sclerostin deficiency combined with glucocorticoid excess resulted in sporadic, sudden, unprovoked and nonconvulsive death caused by peracute hemopericardium and cardiac tamponade. The authors speculate that glucocorticoid excess may also result in increased cardiac risk in situations of sclerostin suppression. They suggest that the developmental effect of excessive glucocorticoids and Sost haploinsufficiency, as well as differences in experimental design and analysis, could be potential reasons for the inconsistency between this study and studies that rescue glucocorticoid induced bone loss through Sost deletion and antisclerostin antibody treatment (Javaheri et al. 2019). However, a link from this preclinical study to the clinical situation has not been demonstrated.

Sclerostin and the immune system

Many signalling pathways, including the Wnt/β-catenin pathway, contribute to regulation of haematopoiesis in adults (Geest & Coffer 2009, Luis et al. 2011, Huang et al. 2012). Sclerostin depletion has been shown to affect B-lymphopoiesis and myelopoiesis, as well as other changes within the bone marrow cavity that could affect haematopoiesis (Donham et al. 2021, Sun et al. 2021). Furthermore, lack of sclerostin results in increased B-cell apoptosis and decreased CXCL12 (an important B-cell growth stimulating factor) expression (Cain et al. 2012). Impaired B-lymphocyte survival causes a reduced number of B-lymphocytes in Sost−/− (MGI:3797839) bone marrow, an effect that is mediated in an indirect cell-extrinsic manner. Conversely, B-lymphocyte development was not affected by deletion of Sost (using Dmp1-Cre) in osteocytes; however, decreased B-cell precursors and immature B-cell subsets were observed when Sost was deleted (using Prx1-Cre) in mesenchymal stem cells (MSCs) (Yee et al. 2018). However, this decrease was not as pronounced as in global Sost−/− mice, suggesting that another sclerostin expressing cell is involved in B-cell development (Cain et al. 2012, Yee et al. 2018). You et al. demonstrated that sclerostin is necessary for inducing T helper 17 (Th17) cell differentiation, which are responsible for bone resorption by promoting the levels of interleukin (IL)-6 and transforming growth factor (TGF)-β. In addition, it has been shown that sclerostin inhibits the differentiation of regulatory T (Treg) cells by reducing the expression of IL-10 and Foxp3, which play essential roles in Treg cell development (You et al. 2018). These findings suggest that further research is warranted to determine whether sclerostin-depleting therapies could result in similar alterations in B-cell dynamics.

Sclerostin and other organs

Muscle

Muscle and bone are both endocrine target tissues and endocrine organs (Brunetti et al. 2017, Giudice & Taylor 2017). Previously it was believed that locomotion was the singular method of crosstalk between muscle and bone. However, crosstalk also occurs when muscle and bone interact with each other through paracrine and endocrine signals, which play key roles in modulation of their mutual development and function (Brotto & Bonewald 2015, Tagliaferri et al. 2015). Many tissue-specific factors released by osteoblasts and osteocytes, such as osteocalcin and insulin-like growth factor 1 (IGF1), appear to have a potential impact on skeletal muscle. Similarly, a number of factors released by muscle with bone-modulating properties have been identified (Magarò et al. 2021). Skeletal muscle cells (myocytes) release myokines, a panel of proteins and cytokines which exert paracrine or endocrine regulatory functions on bone, among other distant organs and tissues (Gomarasca et al. 2020). Myogenic media from differentiating C2C12 myoblast cells altered the functional maturation of osteoblasts and was shown to have inhibitory effects on bone cell differentiation. Sclerostin was identified as a myokine expressed by C2C12 muscle cells and primary murine myoblasts (from C57BL/6J mice) in all stages of differentiation and was dynamically secreted in the myogenic medium during myogenic lineage progression (Magarò et al. 2021). It appears that myoblast sclerostin secretion does not affect circulating sclerostin levels and myokines may affect adjacent bones through a paracrine mechanism, relying on their diffusion across muscle and bone tissues. This suggests that sclerostin released by bone might synergise with sclerostin released by skeletal muscle to inhibit osteogenesis (Magarò et al. 2021). Magarò et al. showed that both muscle- and bone-secreted sclerostin reduce bone formation. Overexpression of sclerostin by myocytes induced trabecular bone loss, but did not influence cortical bone parameters. Interestingly, sclerostin has been found to be expressed by metastatic breast cancer cells. Treating cancer bearing mice with anti-sclerostin (setrusumab) decreased osteolytic bone destruction and muscle weakness (Hesse et al. 2019). Muscle fibre atrophy was reversed by inhibiting the osteoclast-mediated increase in TGF-β1, suggesting cross talk between bone and muscle (Hesse et al. 2019). Although existing evidence shows that sclerostin secreted by both bone and skeletal muscles regulates bone homeostasis, further research is required to fully uncover the processes underlying sclerostin-mediated regulation of bone–muscle interaction. Understanding of these processes could be used to develop innovative therapeutics for diseases such as osteoporosis and sarcopenia.

Liver

Increasing evidence exists concerning the relationship between liver and bone metabolism. Sclerostin is known to be involved in metabolic abnormalities and is possibly increased in patients with impaired glucose regulation, correlating with insulin resistance in skeletal muscle, adipose tissue and the liver (Daniele et al. 2015). Remarkably, higher sclerostin levels are seen in type II diabetes as well as in men with excessive alcohol use (Gennari et al. 2012, Napoli et al. 2018, Martín González et al. 2022). Circulating sclerostin levels were lower in non-alcoholic fatty liver disease (NAFLD) patients and were negatively correlated with multiple metabolic parameters whilst showing no significant correlation to controls (Zou et al. 2020). Evidence suggests that NAFLD is a multisystem disease that affects several organ systems other than the liver and interacts with the regulation of multiple metabolic, endocrine, and pro-inflammatory pathways (Younossi et al. 2018). Significant differences in whole-body or lumbar BMD Z scores between children or adolescents with and without NAFLD have been described, suggesting that liver–bone interaction and the underlying mechanism should be further investigated (Mantovani et al. 2019). Zhou et al. observed reduced bone mass as well as lower sclerostin expression levels in the bone and liver tissues of mice fed on a high-fat diet. These results suggest that liver–lipid–bone interactions may play a key role in the abnormal bone metabolism in NAFLD (Zhou et al. 2021). These studies focused primarily on serum sclerostin levels, sclerostin mRNA expression and changes to bone parameters, thus further research is needed to clarify any effects of sclerostin on liver biology and function and to understand the link between liver and bone cells.

Kidney

Serum sclerostin is increased in patients with chronic kidney disease (CKD), with sclerostin hypothesised to originate from the skeleton and vasculature (Zhu et al. 2011, Brandenburg et al. 2019). The increase in sclerostin with CKD progression is most notable in stage 3 CKD, indicating that sclerostin is inversely proportional to glomerular filtration rate (Pelletier et al. 2015). The heightened sclerostin expression seen in CKD patients is thought to be a result of many factors, including sclerostin renal retention (Sabbagh et al. 2012, Bruzzese et al. 2016). However, it has been reported that urinary sclerostin excretion increases simultaneously with declining estimated glomerular filtration rate (eGFR) and increasing sclerostin secretion from osteocytes (Sabbagh et al. 2012, Bruzzese et al. 2016). The importance of sclerostin for cardiovascular and bone health in CKD patients is not fully understood. Studies have shown that CKD patients are at a higher risk of CVEs. Additionally, sclerostin is expressed within their vasculature with highly active vascular calcification processes. Within this patient population, anti-sclerostin antibody treatments have a weakened efficacy on bone, coupled with an increased rate of hypocalcaemia (Cejka 2021). The function of sclerostin in the uremic arteries is unknown; however, it may be postulated that its expression within the vasculature could be a marker for uremic damage (Marchand et al. 2011). In patients undergoing peritoneal dialysis, low serum sclerostin was related to an increased survival rate as well as a lower chance of a CVE. This relationship does not correlate with haemodialysis patients (Zou et al. 2020). Sclerostin may have utility as a clinically relevant marker of disturbed bone metabolism in end-stage kidney disease patients (De Maré et al. 2019b). However, further in vitro and in vivo cause–effect studies are required to better understand the role of sclerostin in the kidney and how it relates to both bone and vasculature.

Sclerostin as a therapeutic

An increased understanding of sclerostin and its function in bone biology has resulted in the investigation of sclerostin as a therapeutic. Here, we focus on the latest advances in sclerostin as a treatment for sclerosteosis, OA and osteosarcoma.

Sclerosteosis

Sclerostin replacement therapy has recently been investigated as a potential treatment for sclerosteosis by replacing absent sclerostin with recombinant wildtype or sclerostin fusion constructs (Dreyer et al. 2021). Dreyer et al. demonstrated that wildtype murine sclerostin, sclerostin human immunoglobulin G1 (IgG1) fragment crystallisable (Fc) fusion protein (mScl-hFc) and sclerostin human Fc fusion with a bone targeting C-terminal poly-aspartate motif (mScl-hFc-PD) bound with high affinity to the extracellular domain of the LRP6 Wnt co-receptor and inhibited mineralisation in a murine osteoblast-like cell line. Fusing sclerostin with IgG1 human Fc increased protein half-life from minutes to longer than a day in WT and Sost –/– mice. Modest but significant reductions in trabecular volumetric bone mineral density (vBMD) and bone volume fraction (BV/TV) of 20% and 15%, respectively, were observed after 6 weeks in Sost –/– mice treated with mScl-hFc-PD. It is thus possible that the bone-targeting moiety may have been beneficial. Cortical bone and bone formation markers remained unchanged and anti-sclerostin antibodies were observed. Although these antibodies are the most likely cause of modest efficacy, increased DKK1 concentrations have been reported in mice and rats treated with anti-sclerostin antibodies (Taylor et al. 2016, Holdsworth et al. 2018). It is possible that increased DKK1 levels are also present in the Sost–/– mice and potentially contributed to the modest effects from sclerostin constructs (Dreyer et al. 2021). This study indicates that a protein replacement approach might not be effective for long-term use and alternatives, such as small molecules, should be considered.

Osteoarthritis

Suppression of Wnt signalling by upregulated SOST/sclerostin in cartilage derived from biopsies of OA patients and elevated sclerostin in traumatic OA models suggest that sclerostin might play a key protective role in the maintenance of articular homeostasis (Karlsson et al. 2010, Chan et al. 2011). Indeed, Chang et al. found that sclerostin inhibits cartilage degradation after traumatic injury by downregulating catabolic enzymes/activity of proteolytic enzymes, such as matrix metalloproteinases (MMP and MMP2/3), in osteopenic SOST transgenic mice (Chang et al. 2018). MMP activity was also significantly decreased in both SOST transgenic and Sost knockout mice after intra-articular administration of recombinant sclerostin, immediately after joint injury. These findings suggest that elevated levels of sclerostin immediately post-injury can aid the joint in maintaining its articular cartilage integrity in post-traumatic settings (Chang et al. 2018). In addition, sclerostin overexpression may protect OA joints from excessive osteophyte formation, whilst lack of sclerostin could protect the femur from bone loss that results from disuse or injury (Chang et al. 2018). Sclerostin treatment of ATDC5 cells, a well-established in vitro model of chondrogenesis, promoted chondrogenic gene expression and suppressed hypertrophic differentiation (Atsumi et al. 1990, Yamaguchi et al. 2018). Moreover, addition of sclerostin following IL-1β treatment repressed the upregulation of Wnt/β-catenin signalling and inhibited progression of chondrogenic differentiation and terminal calcification promoted by IL-1β addition to ATDC5 cells (Miyatake et al. 2020). As with the Chang et al. study, these results suggest that suppression of Wnt signalling by sclerostin might be key for the maintenance of articular homeostasis. Furthermore, treatment with the anti-malarial drug dihydroartemisinin has been shown to enhance sclerostin expression and subsequently reduce subchondral bone remodelling in a surgically induced OA mouse model (Ma et al. 2021). Interestingly, sclerostin has been shown to be upregulated by OA chondrocytes in end-stage disease, which may be an attempt to prevent further destruction by slowing down Wnt-driven catabolic responses (Chan et al. 2011). There is evidence that sclerostin modulates cartilage homeostasis and may be of therapeutic benefit in early stage disease; however, its application as a sole therapy for advanced disease is likely insufficient to reverse the plethora of pathological structural changes observed.

Osteosarcoma

Osteosarcoma has one of the lowest survival rates of all paediatric cancers as it is highly resistant to treatment, partially due to its highly diverse and heterogeneous nature (Gill & Gorlick 2021). New treatments are thus urgently needed. The Wnt/β-catenin signalling pathway may play an important role in osteosarcoma tumourogenesis (Inagaki et al. 2016). Indeed, Ideta et al. recently reported that sclerostin inhibits the Wnt/β-catenin pathway as well as proliferation and migration in both murine (LM8) and human (143B) osteosarcoma cell lines (Ideta et al. 2021). Sclerostin also inhibited tumour growth in mice with transplanted osteosarcoma cells, extending overall survival rate. SOST expression was significantly decreased in tumour-bearing bones from 15-week-old osteoblast-specific Wntless (Wls) loss-of-function OS mice (WlsΔOB-OS mice) (Matsuoka et al. 2020). These results corroborate the findings by Zou et al. that SOST gene silencing activates the Wnt/β-catenin pathway, resulting in decreased apoptosis, increased proliferation, and invasion and migration of osteosarcoma cells collected from primary tumour tissues of osteosarcoma patients (Zou et al. 2017). Taken together, these findings suggest that local treatment with drugs that increase local sclerostin concentration or reduce Wnt expression/activity by mimicking sclerostin function could be effective for the treatment of osteosarcoma whilst avoiding systemic negative impacts on bone.

Future

As discussed in this review, sclerostin has a potential role in regulating and maintaining homeostasis in many organs beyond that of the bone, with associations to the immune system, kidney, liver, muscle, cartilage and cardiovascular system. However, it is important to note that regulated expression is essential to ensure controlled balance of canonical Wnt signalling. Many reports suggest that an imbalance in sclerostin expression/function may be linked to metabolic diseases, musculoskeletal disorders and cardiovascular events. However, it is not yet understood whether dysregulation of sclerostin is a causative event in all these conditions, as observed in sclerosteosis, or whether sclerostin expression is affected during disease progression. Nevertheless, sclerostin levels are altered and more research (e.g. mechanistic in vivo) is required to underpin its role and aid therapeutic development. Research is well underway regarding the modulation of sclerostin function as a therapeutic approach, with romosozumab for post-menopausal osteoporosis already approved by multiple regulators. However, as seen with romosozumab, changes in bone mineral density might become smaller following prolonged treatment with canonical Wnt-signalling pathway targeting therapeutics (McClung et al. 2018). Novel approaches including sclerostin replacement therapy, small molecule intervention, epigenetic modulation and aptamer targeting of sclerostin may shed light on the future of sclerostin-based therapeutics and offer exciting avenues in novel drug development approaches.

Declaration of interest

SR is a previous employee of UCB Pharma and is in receipt of research funding from UCB Pharma to investigate sclerostin biology in the context of Sclerosteosis. TD, JK and LW have no conflicts of interest that could be perceived as prejudicing the impartiality of this review.

Funding

This work did not receive any specific grant from any funding agency in the public, commercial or not-for-profit sector.

References

  • Ai M, Holmen SL, Van Hul W, Williams BO & & Warman ML 2005 Reduced affinity to and inhibition by DKK1 form a common mechanism by which high bone mass-associated missense mutations in LRP5 affect canonical Wnt signaling. Molecular and Cellular Biology 25 49464955. (https://doi.org/10.1128/MCB.25.12.4946-4955.2005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Akkiraju H & & Nohe A 2015 Role of chondrocytes in cartilage formation, progression of osteoarthritis and cartilage regeneration. Journal of Developmental Biology 3 177192. (https://doi.org/10.3390/jdb3040177)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Allison H, Holdsworth G & & Mcnamara LM 2020 Scl-Ab reverts pro-osteoclastogenic signalling and resorption in estrogen deficient osteocytes. BMC Molecular and Cell Biology 21 78. (https://doi.org/10.1186/s12860-020-00322-w)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Atkins GJ, Rowe PS, Lim HP, Welldon KJ, Ormsby R, Wijenayaka AR, Zelenchuk L, Evdokiou A & & Findlay DM 2011 Sclerostin is a locally acting regulator of late-osteoblast/preosteocyte differentiation and regulates mineralization through a MEPE-ASARM-dependent mechanism. Journal of Bone and Mineral Research 26 14251436. (https://doi.org/10.1002/jbmr.345)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Atkinson EG & & Delgado‐Calle J 2019 The emerging role of osteocytes in cancer in bone. JBMR Plus 3 e10186. (https://doi.org/10.1002/jbm4.10186)

  • Atsumi T, Miwa Y, Kimata K & & Ikawa Y 1990 A chondrogenic cell line derived from a differentiating culture of AT805 teratocarcinoma cells. Cell Differentiation and Development 30 109116. (https://doi.org/10.1016/0922-3371(9090079-c)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 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.2001 Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST). Human Molecular Genetics 10 537543. (https://doi.org/10.1093/hmg/10.5.537)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Balemans W, Patel N, Ebeling M, Van Hul E, Wuyts W, Lacza C, Dioszegi M, Dikkers FG, Hildering P, Willems PJ, et al.2002 Identification of a 52 kb deletion downstream of the SOST gene in patients with van Buchem disease. Journal of Medical Genetics 39 9197. (https://doi.org/10.1136/jmg.39.2.91)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Balemans W, Piters E, Cleiren E, Ai M, Van Wesenbeeck L, Warman ML & & Van Hul W 2008 The binding between sclerostin and LRP5 is altered by DKK1 and by high-bone mass LRP5 mutations. Calcified Tissue International 82 445453. (https://doi.org/10.1007/s00223-008-9130-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baron R, Ferrari S & & Russell RGG 2011 Denosumab and bisphosphonates: different mechanisms of action and effects. Bone 48 677692. (https://doi.org/10.1016/j.bone.2010.11.020)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Beighton P 1988 Sclerosteosis. Journal of Medical Genetics 25 200203. (https://doi.org/10.1136/jmg.25.3.200)

  • Bovijn J, Krebs K, Chen CY, Boxall R, Censin JC, Ferreira T, Pulit SL, Glastonbury CA, Laber S, Millwood IY, et al.2020 Evaluating the cardiovascular safety of sclerostin inhibition using evidence from meta-analysis of clinical trials and human genetics. Science Translational Medicine 12 549. (https://doi.org/10.1126/scitranslmed.aay6570)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bovijn J, Krebs K, Chen CY, Boxall R, Censin JC, Ferreira T, Pulit SL, Glastonbury CA, Laber S, Millwood IY, et al.2021 Response to comment on “Evaluating the cardiovascular safety of sclerostin inhibition using evidence from meta-analysis of clinical trials and human genetics”. Science Translational Medicine 13 eabf4530. (https://doi.org/10.1126/scitranslmed.abf4530)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brandenburg VM, Verhulst A, Babler A, D'haese PC, Evenepoel P & & Kaesler N 2019 Sclerostin in chronic kidney disease–mineral bone disorder think first before you block it! Nephrology, Dialysis, Transplantation 34 408414. (https://doi.org/10.1093/ndt/gfy129)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brotto M & & Bonewald L 2015 Bone and muscle: interactions beyond mechanical. Bone 80 109114. (https://doi.org/10.1016/j.bone.2015.02.010)

  • Brown JP, Prince RL, Deal C, Recker RR, Kiel DP, De Gregorio LH, Hadji P, Hofbauer LC, Álvaro-Gracia JM, Wang H, et al.2009 Comparison of the Effect of Denosumab and Alendronate on BMD and Biochemical Markers of Bone Turnover in Postmenopausal Women With Low Bone Mass: a Randomized, Blinded, Phase 3 Trial. Journal of Bone and Mineral Research 24 153161. (https://doi.org/10.1359/jbmr.0809010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brunetti G, D’amelio P, Wasniewska M, Mori G & & Faienza MF 2017. Editorial Editorial: Bone: endocrine target and organ. Frontiers in Endocrinology 8 354. (https://doi.org/10.3389/fendo.2017.00354)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brunetti G, Oranger A, Mori G, Specchia G, Rinaldi E, Curci P, Zallone A, Rizzi R, Grano M & & Colucci S 2011 Sclerostin is overexpressed by plasma cells from multiple myeloma patients. Annals of the New York Academy of Sciences 1237 1923. (https://doi.org/10.1111/j.1749-6632.2011.06196.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bruzzese A, Lacquaniti A, Cernaro V, Ricciardi CA, Loddo S, Romeo A, Montalto G, Costantino G, Torre F, Pettinato G, et al.2016 Sclerostin levels in uremic patients: a link between bone and vascular disease. Renal Failure 38 759764. (https://doi.org/10.3109/0886022X.2016.1160207)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bullock WA, Hoggatt AM, Horan DJ, Elmendorf AJ, Sato AY, Bellido T, Loots GG, Pavalko FM & & Robling AG 2019 Lrp4 mediates bone homeostasis and mechanotransduction through interaction with sclerostin in vivo. iScience 20 205215. (https://doi.org/10.1016/j.isci.2019.09.023)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buniello A, Macarthur JAL, Cerezo M, Harris LW, Hayhurst J, Malangone C, Mcmahon A, Morales J, Mountjoy E, Sollis E, et al.2019 The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Research 47 D1005D1012. (https://doi.org/10.1093/nar/gky1120)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cain CJ, Rueda R, Mclelland B, Collette NM, Loots GG & & Manilay JO 2012 Absence of sclerostin adversely affects B cell survival. Journal of Bone and Mineral Research 27 14511461. (https://doi.org/10.1002/jbmr.1608)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Capulli M, Paone R & & Rucci N 2014 Osteoblast and osteocyte: games without frontiers. Archives of Biochemistry and Biophysics 561 312. (https://doi.org/10.1016/j.abb.2014.05.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Catalano A, Bellone F, Morabito N & & Corica F 2020 Sclerostin and vascular pathophysiology. International Journal of Molecular Sciences 21 4779. (https://doi.org/10.3390/ijms21134779)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cejka D 2021 Cardiovascular safety of anti-sclerostin therapy in chronic kidney disease. Metabolites 11 770. (https://doi.org/10.3390/metabo11110770)

  • Chan BY, Fuller ES, Russell AK, Smith SM, Smith MM, Jackson MT, Cake MA, Read RA, Bateman JF, Sambrook PN, et al.2011 Increased chondrocyte sclerostin may protect against cartilage degradation in osteoarthritis. Osteoarthritis and Cartilage 19 874885. (https://doi.org/10.1016/j.joca.2011.04.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chang JC, Christiansen BA, Murugesh DK, Sebastian A, Hum NR, Collette NM, Hatsell S, Economides AN, Blanchette CD & & Loots GG 2018 SOST/sclerostin improves posttraumatic osteoarthritis and inhibits MMP2/3 expression after injury. Journal of Bone and Mineral Research 33 11051113. (https://doi.org/10.1002/jbmr.3397)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chavassieux PM, Arlot ME, Reda C, Wei L, Yates AJ & & Meunier PJ 1997 Histomorphometric assessment of the long-term effects of alendronate on bone quality and remodeling in patients with osteoporosis. Journal of Clinical Investigation 100 14751480. (https://doi.org/10.1172/JCI119668)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cheng JCY, Tang NLS, Yeung HY & & Miller N 2007 Genetic association of complex traits: using idiopathic scoliosis as an example. Clinical Orthopaedics and Related Research 462 3844. (https://doi.org/10.1097/BLO.0b013e3180d09dcc)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Choi HY, Dieckmann M, Herz J & & Niemeier A 2009 Lrp4, a novel receptor for Dickkopf 1 and sclerostin, is expressed by osteoblasts and regulates bone growth and turnover in vivo. PLoS One 4 e7930. (https://doi.org/10.1371/journal.pone.0007930)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Colucci S, Brunetti G, Oranger A, Mori G, Sardone F, Specchia G, Rinaldi E, Curci P, Liso V, Passeri G, et al.2011 Myeloma cells suppress osteoblasts through sclerostin secretion. Blood Cancer Journal 1 e27. (https://doi.org/10.1038/bcj.2011.22)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S, Hofbauer LC, Lau E, Lewiecki EM, Miyauchi A, et al.2016 Romosozumab treatment in postmenopausal women with osteoporosis. New England Journal of Medicine 375 15321543. (https://doi.org/10.1056/NEJMoa1607948)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Daniele G, Winnier D, Mari A, Bruder J, Fourcaudot M, Pengou Z, Tripathy D, Jenkinson C & & Folli F 2015 Sclerostin and insulin resistance in prediabetes: evidence of a cross talk between bone and glucose metabolism. Diabetes Care 38 15091517. (https://doi.org/10.2337/dc14-2989)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • De Maré A, Maudsley S, Azmi A, Hendrickx JO, Opdebeeck B, Neven E, D’haese PC & & Verhulst A 2019a Sclerostin as regulatory molecule in vascular media calcification and the bone–vascular axis. Toxins 11 428. (https://doi.org/10.3390/toxins11070428)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • De Maré A, Opdebeeck B, Neven E, D'haese PC & & Verhulst A 2022 Sclerostin protects against vascular calcification development in mice. Journal of Bone and Mineral Research 37 687699. (https://doi.org/10.1002/jbmr.4503)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • De Maré A, Verhulst A, Cavalier E, Delanaye P, Behets GJ, Meijers B, Kuypers D, D’haese PC & & Evenepoel P 2019b Clinical inference of serum and bone sclerostin levels in patients with end-stage kidney disease. Journal of Clinical Medicine 8 2027. (https://doi.org/10.3390/jcm8122027)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Delgado-Calle J, Anderson J, Cregor MD, Condon KW, Kuhstoss SA, Plotkin LI, Bellido T & & Roodman GD 2017 Genetic deletion of Sost or pharmacological inhibition of sclerostin prevent multiple myeloma-induced bone disease without affecting tumor growth. Leukemia 31 26862694. (https://doi.org/10.1038/leu.2017.152)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Delgado-Calle J, Anderson J, Cregor MD, Hiasa M, Chirgwin JM, Carlesso N, Yoneda T, Mohammad KS, Plotkin LI, Roodman GD, et al.2016 Bidirectional Notch signaling and osteocyte-derived factors in the bone marrow microenvironment promote tumor cell proliferation and bone destruction in multiple myeloma. Cancer Research 76 10891100. (https://doi.org/10.1158/0008-5472.CAN-15-1703)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Delgado-Calle J, Bellido T & & Roodman GD 2014 Role of osteocytes in multiple myeloma bone disease. Current Opinion in Supportive and Palliative Care 8 407–413. (https://doi.org/10.1097/SPC.0000000000000090)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Desjardins L, Liabeuf S, Oliveira RB, Louvet L, Kamel S, Lemke HD, Vanholder R, Choukroun G, Massy ZA & European Uremic Toxin (EUTox) Work Group 2014 Uremic toxicity and sclerostin in chronic kidney disease patients. Nephrologie et Therapeutique 10 463470. (https://doi.org/10.1016/j.nephro.2014.04.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Didangelos A, Yin X, Mandal K, Saje A, Smith A, Xu Q, Jahangiri M & & Mayr M 2011 Extracellular matrix composition and remodeling in human abdominal aortic aneurysms: a proteomics approach. Molecular and Cellular Proteomics 10 M111.008128. (https://doi.org/10.1074/mcp.M111.008128)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dobbs MB, Buckwalter J & & Saltzman C 1999 Osteoporosis: The increasing role of the orthopaedist. Iowa Orthopaedic Journal 19 4352.

  • Donham C, Chicana B, Robling AG, Mohamed A, Elizaldi S, Chi M, Freeman B, Millan A, Murugesh DK, Hum NR, et al.2021 Sclerostin depletion induces inflammation in the bone marrow of mice. International Journal of Molecular Sciences 22 9111. (https://doi.org/10.3390/ijms22179111)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dreyer T, Shah M, Doyle C, Greenslade K, Penney M, Creeke P, Kotian A, Ke HZ, Naidoo V & & Holdsworth G 2021 Recombinant sclerostin inhibits bone formation in vitro and in a mouse model of sclerosteosis. Journal of Orthopaedic Translation 29 134–142. (https://doi.org/10.1016/j.jot.2021.05.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eastell R, Rosen CJ, Black DM, Cheung AM, Murad MH & & Shoback D 2019 Pharmacological management of osteoporosis in postmenopausal women: an Endocrine Society* clinical practice guideline. Journal of Clinical Endocrinology and Metabolism 104 15951622. (https://doi.org/10.1210/jc.2019-00221)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eda H, Santo L, Wein MN, Hu DZ, Cirstea DD, Nemani N, Tai YT, Raines SE, Kuhstoss SA, Munshi NC, et al.2016 Regulation of sclerostin expression in multiple myeloma by Dkk-1; a potential therapeutic strategy for myeloma bone disease. Journal of Bone and Mineral Research: The Official Journal of the American Society for Bone and Mineral Research 31 12251234. (https://doi.org/10.1002/jbmr.2789)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fijalkowski I, Geets E, Steenackers E, Van Hoof V, Ramos FJ, Mortier G, Fortuna AM, Van Hul W & & Boudin E 2016 A novel domain-specific mutation in a sclerosteosis patient suggests a role of LRP4 as an anchor for sclerostin in human bone. Journal of Bone and Mineral Research 31 874881. (https://doi.org/10.1002/jbmr.2782)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Florio M, Gunasekaran K, Stolina M, Li X, Liu L, Tipton B, Salimi-Moosavi H, Asuncion FJ, Li C, Sun B, et al.2016 A bispecific antibody targeting sclerostin and DKK-1 promotes bone mass accrual and fracture repair. Nature Communications 7 11505. (https://doi.org/10.1038/ncomms11505)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fuggle NR, Cooper C, Harvey NC, Al-Daghri N, Brandi ML, Bruyere O, Cano A, Dennison EM, Diez-Perez A, Kaufman JM, et al.2020 Assessment of cardiovascular safety of anti-osteoporosis drugs. Drugs 80 15371552. (https://doi.org/10.1007/s40265-020-01364-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gau YC, Yeh TJ, Hsu CM, Hsiao SY & & Hsiao HH 2022 Pathogenesis and treatment of myeloma-related bone disease. International Journal of Molecular Sciences 23 3112. (https://doi.org/10.3390/ijms23063112)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaudio A, Fiore V, Rapisarda R, Sidoti MH, Xourafa A, Catalano A, Tringali G, Zanoli L, Signorelli SS & & Fiore CE 2017 Sclerostin is a possible candidate marker of arterial stiffness: results from a cohort study in Catania. Molecular Medicine Reports 15 34203424. (https://doi.org/10.3892/mmr.2017.6390)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gaudio A, Privitera F, Pulvirenti I, Canzonieri E, Rapisarda R & & Fiore CE 2014 The relationship between inhibitors of the Wnt signalling pathway (sclerostin and Dickkopf-1) and carotid intima-media thickness in postmenopausal women with type 2 diabetes mellitus. Diabetes and Vascular Disease Research 11 4852. (https://doi.org/10.1177/1479164113510923)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Geest CR & & Coffer PJ 2009 MAPK signaling pathways in the regulation of hematopoiesis. Journal of Leukocyte Biology 86 237250. (https://doi.org/10.1189/jlb.0209097)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gennari L, Merlotti D, Valenti R, Ceccarelli E, Ruvio M, Pietrini MG, Capodarca C, Franci MB, Campagna MS, Calabro A, et al.2012 Circulating sclerostin levels and bone turnover in type 1 and type 2 diabetes. Journal of Clinical Endocrinology and Metabolism 97 17371744. (https://doi.org/10.1210/jc.2011-2958)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gill J & & Gorlick R 2021 Advancing therapy for osteosarcoma. Nature Reviews. Clinical Oncology 18 609624. (https://doi.org/10.1038/s41571-021-00519-8)

  • Giudice J & & Taylor JM 2017 Muscle as a paracrine and endocrine organ. Current Opinion in Pharmacology 34 4955. (https://doi.org/10.1016/j.coph.2017.05.005)

  • Giuliani N, Ferretti M, Bolzoni M, Storti P, Lazzaretti M, Dalla Palma B, Bonomini S, Martella E, Agnelli L, Neri A, et al.2012 Increased osteocyte death in multiple myeloma patients: role in myeloma-induced osteoclast formation. Leukemia 26 13911401. (https://doi.org/10.1038/leu.2011.381)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Glass DA, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers H, Taketo MM, Long F, Mcmahon AP, Lang RA, et al.2005 Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Developmental Cell 8 751764. (https://doi.org/10.1016/j.devcel.2005.02.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Glorieux FH, Devogelaer JP, Durigova M, Goemaere S, Hemsley S, Jakob F, Junker U, Ruckle J, Seefried L & & Winkle PJ 2017 BPS804 Anti-Sclerostin antibody in Adults with Moderate Osteogenesis imperfecta: results of a Randomized Phase 2a Trial. Journal of Bone and Mineral Research 32 14961504. (https://doi.org/10.1002/jbmr.3143)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Goldring MB 2012 Chondrogenesis, chondrocyte differentiation, and articular cartilage metabolism in health and osteoarthritis. Therapeutic Advances in Musculoskeletal Disease 4 269285. (https://doi.org/10.1177/1759720X12448454)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gomarasca M, Banfi G & & Lombardi G 2020 Myokines: the endocrine coupling of skeletal muscle and bone. Advances in Clinical Chemistry 94 155218. (https://doi.org/10.1016/bs.acc.2019.07.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gorter EA, Reinders CR, Krijnen P, Appelman-Dijkstra NM & & Schipper IB 2022 Serum sclerostin levels in osteoporotic fracture patients. European Journal of Trauma and Emergency Surgery 48 48574865. (https://doi.org/10.1007/s00068-022-02017-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Habibi H, Abroun S, Hajifathali A, Soleimani M, Kaviani S, Kalantari N & & Eslahchi S 2013 Osteogenic inhibition in multiple myeloma. Cell Journal 15 266271.

  • Hesse E, Schröder S, Brandt D, Pamperin J, Saito H & & Taipaleenmäki H 2019 Sclerostin inhibition alleviates breast cancer–induced bone metastases and muscle weakness. JCI Insight 5 e125543. (https://doi.org/10.1172/jci.insight.125543)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holdsworth G, Greenslade K, Jose J, Stencel Z, Kirby H, Moore A, Ke HZ & & Robinson MK 2018 Dampening of the bone formation response following repeat dosing with sclerostin antibody in mice is associated with up-regulation of Wnt antagonists. Bone 107 93103. (https://doi.org/10.1016/j.bone.2017.11.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holdsworth G, Roberts SJ & & Ke HZ 2019 Novel actions of sclerostin on bone. Journal of Molecular Endocrinology 62 R167R185. (https://doi.org/10.1530/JME-18-0176)

  • Holdsworth G, Slocombe P, Doyle C, Sweeney B, Veverka V, Le Riche K, Franklin RJ, Compson J, Brookings D, Turner J, et al.2012 Characterization of the interaction of sclerostin with the low density lipoprotein receptor-related protein (LRP) family of Wnt co-receptors. Journal of Biological Chemistry 287 2646426477. (https://doi.org/10.1074/jbc.M112.350108)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holdsworth G, Staley JR, Hall P, Van Koeverden I, Vangjeli C, Okoye R, Boyce RW, Turk JR, Armstrong M, Wolfreys A, et al.2021 Sclerostin downregulation globally by naturally occurring genetic variants, or locally in atherosclerotic plaques, does not associate with cardiovascular events in humans. Journal of Bone and Mineral Research 36 13261339. (https://doi.org/10.1002/jbmr.4287)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Holm H, Sulem P, Tragante V, Thorsteinsdottir U, Gudbjartsson DF & & Stefansson K 2021 Comment on “Evaluating the cardiovascular safety of sclerostin inhibition using evidence from meta-analysis of clinical trials and human genetics”. Science Translational Medicine 13 eabe8497. (https://doi.org/10.1126/scitranslmed.abe8497)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hong AR, Yang JY, Lee JY, Suh J, Lee YS, Kim JE & & Kim SW 2022 Reactivation of bone lining cells are attenuated over repeated anti-sclerostin antibody administration. Calcified Tissue International 111 495505. (https://doi.org/10.1007/s00223-022-01013-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hsu BG, Liou HH, Lee CJ, Chen YC, Ho GJ & & Lee MC 2016 Serum sclerostin as an independent marker of peripheral arterial stiffness in renal transplantation recipients: a cross-sectional study. Medicine (Baltimore) 95 e3300. (https://doi.org/10.1097/MD.0000000000003300)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Huang J, Nguyen-McCarty M, Hexner EO, Danet-Desnoyers G & & Klein PS 2012 Maintenance of hematopoietic stem cells through regulation of Wnt and mTOR pathways. Nature Medicine 18 17781785. (https://doi.org/10.1038/nm.2984)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ideta H, Yoshida K, Okamoto M, Sasaki J, Kito M, Aoki K, Yoshimura Y, Suzuki S, Tanaka A, Takazawa A, et al.2021 Antitumor effect of sclerostin against osteosarcoma. Cancers 13 6015. (https://doi.org/10.3390/cancers13236015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Inagaki Y, Hookway ES, Kashima TG, Munemoto M, Tanaka Y, Hassan AB, Oppermann U & & Athanasou NA 2016 Sclerostin expression in bone tumours and tumour-like lesions. Histopathology 69 470478. (https://doi.org/10.1111/his.12953)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Javaheri B, Herbert E, Hopkinson M, Al-Jazzar A & & Pitsillides AA 2019 Sost haploinsufficiency provokes peracute lethal cardiac tamponade without rescuing the osteopenia in a mouse model of excess glucocorticoids. American Journal of Pathology 189 753761. (https://doi.org/10.1016/j.ajpath.2018.12.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Karlsson C, Dehne T, Lindahl A, Brittberg M, Pruss A, Sittinger M & & Ringe J 2010 Genome-wide expression profiling reveals new candidate genes associated with osteoarthritis. Osteoarthritis and Cartilage 18 581592. (https://doi.org/10.1016/j.joca.2009.12.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ke HZ, Richards WG, Li X & & Ominsky MS 2012 Sclerostin and Dickkopf-1 as therapeutic targets in bone diseases. Endocrine Reviews 33 747783. (https://doi.org/10.1210/er.2011-1060)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim DH, Rogers JR, Fulchino LA, Kim CA, Solomon DH & & Kim SC 2015 Bisphosphonates and risk of cardiovascular events: a meta-analysis. PLoS One 10 e0122646. (https://doi.org/10.1371/journal.pone.0122646)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim SP, Frey JL, Li Z, Kushwaha P, Zoch ML, Tomlinson RE, Da H, Aja S, Noh HL, Kim JK, et al.2017a Sclerostin influences body composition by regulating catabolic and anabolic metabolism in adipocytes. PNAS 114 E11238E11247. (https://doi.org/10.1073/pnas.1707876115)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kim SW, Lu Y, Williams EA, Lai F, Lee JY, Enishi T, Balani DH, Ominsky MS, Ke HZ, Kronenberg HM, et al.2017b Sclerostin antibody administration converts bone lining cells into active osteoblasts. Journal of Bone and Mineral Research 32 892901. (https://doi.org/10.1002/jbmr.3038)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Koide M, Yamashita T, Murakami K, Uehara S, Nakamura K, Nakamura M, Matsushita M, Ara T, Yasuda H, Penninger JM, et al.2020 Sclerostin expression in trabecular bone is downregulated by osteoclasts. Scientific Reports 10 13751. (https://doi.org/10.1038/s41598-020-70817-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Koide M, Yamashita T, Nakamura K, Yasuda H, Udagawa N & & Kobayashi Y 2022 Evidence for the major contribution of remodeling-based bone formation in sclerostin-deficient mice. Bone 160 116401. (https://doi.org/10.1016/j.bone.2022.116401)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Koos R, Brandenburg V, Mahnken AH, Schneider R, Dohmen G, Autschbach R, Marx N & & Kramann R 2013 Sclerostin as a potential novel biomarker for aortic valve calcification: an in-vivo and ex-vivo study. Journal of Heart Valve Disease 22 317325.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kranenburg G, Bartstra JW, Weijmans M, De Jong PA, Mali WP, Verhaar HJ, Visseren FLJ & & Spiering W 2016 Bisphosphonates for cardiovascular risk reduction: a systematic review and meta-analysis. Atherosclerosis 252 106115. (https://doi.org/10.1016/j.atherosclerosis.2016.06.039)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Krishna SM, Seto SW, Jose RJ, Li J, Morton SK, Biros E, Wang Y, Nsengiyumva V, Lindeman JH, Loots GG, et al.2017 Wnt signaling pathway inhibitor sclerostin inhibits angiotensin II-induced aortic aneurysm and atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology 37 553566. (https://doi.org/10.1161/ATVBAHA.116.308723)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kuipers AL, Miljkovic I, Carr JJ, Terry JG, Nestlerode CS, Ge Y, Bunker CH, Patrick AL & & Zmuda JM 2015 Association of circulating sclerostin with vascular calcification in Afro-Caribbean men. Atherosclerosis 239 218223. (https://doi.org/10.1016/j.atherosclerosis.2015.01.010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Langdahl BL, Hofbauer LC & & Forfar JC 2021 Cardiovascular safety and sclerostin inhibition. Journal of Clinical Endocrinology and Metabolism 106 18451853. (https://doi.org/10.1210/clinem/dgab193)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lawson MA, Paton-Hough JM, Evans HR, Walker RE, Harris W, Ratnabalan D, Snowden JA & & Chantry AD 2015 NOD/SCID-gamma mice are an ideal strain to assess the efficacy of therapeutic agents used in the treatment of myeloma bone disease. PLoS One 10 e0119546. (https://doi.org/10.1371/journal.pone.0119546)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leder BZ, O'dea LSL, Zanchetta JR, Kumar P, Banks K, Mckay K, Lyttle CR & & Hattersley G 2015 Effects of abaloparatide, a human parathyroid hormone-related peptide analog, on bone mineral density in postmenopausal women with osteoporosis. Journal of Clinical Endocrinology and Metabolism 100 697706. (https://doi.org/10.1210/jc.2014-3718)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lei Y, Fu X, Li P, Lin S, Yan Q, Lai Y, Liu X, Wang Y, Bai X, Liu C, et al.2020 LIM domain proteins Pinch1/2 regulate chondrogenesis and bone mass in mice. Bone Research 8 37. (https://doi.org/10.1038/s41413-020-00108-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leto G, D'onofrio L, Lucantoni F, Zampetti S, Campagna G, Foffi C, Moretti C, Carlone A, Palermo A, Leopizzi M, et al.2019 Sclerostin is expressed in the atherosclerotic plaques of patients who undergoing carotid endarterectomy. Diabetes/Metabolism Research and Reviews 35 e3069. (https://doi.org/10.1002/dmrr.3069)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leupin O, Piters E, Halleux C, Hu S, Kramer I, Morvan F, Bouwmeester T, Schirle M, Bueno-Lozano M, Ramos Fuentes FJ, et al.2011 Bone overgrowth-associated mutations in the LRP4 gene impair sclerostin facilitator function. Journal of Biological Chemistry 286 1948919500. (https://doi.org/10.1074/jbc.M110.190330)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lewiecki EM, Blicharski T, Goemaere S, Lippuner K, Meisner PD, Miller PD, Miyauchi A, Maddox J, Chen L & & Horlait S 2018 A Phase III randomized placebo-controlled trial to evaluate efficacy and safety of Romosozumab in men with osteoporosis. Journal of Clinical Endocrinology and Metabolism 103 31833193. (https://doi.org/10.1210/jc.2017-02163)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li J, Xue J, Jing Y, Wang M, Shu R, Xu H, Xue C, Feng J, Wang P & & Bai D 2019a SOST deficiency aggravates osteoarthritis in mice by promoting sclerosis of subchondral bone. BioMed Research International 2019 7623562. (https://doi.org/10.1155/2019/7623562)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li M, Zhou H, Yang M & & Xing C 2019b Relationship between serum sclerostin, vascular sclerostin expression and vascular calcification assessed by different methods in ESRD patients eligible for renal transplantation: a cross-sectional study. International Urology and Nephrology 51 311323. (https://doi.org/10.1007/s11255-018-2033-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Li X, Ominsky MS, Niu QT, Sun N, Daugherty B, D'agostin D, Kurahara C, Gao Y, Cao J, Gong J, et al.2008 Targeted deletion of the sclerostin gene in mice results in increased bone formation and bone strength. Journal of Bone and Mineral Research 23 860869. (https://doi.org/10.1359/jbmr.080216)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Libby P, Buring JE, Badimon L, Hansson GK, Deanfield J, Bittencourt MS, Tokgozoglu L & & Lewis EF 2019 Atherosclerosis. Nature Reviews. Disease Primers 5 56. (https://doi.org/10.1038/s41572-019-0106-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liberman UA, Weiss SR, Bröll J, Minne HW, Quan H, Bell NH, Rodriguez-Portales J, Downs RW, Dequeker J, Favus M, et al.1995 Effect of oral alendronate on bone mineral density and the incidence of fractures in postmenopausal osteoporosis. The Alendronate Phase III Osteoporosis Treatment Study Group. New England Journal of Medicine 333 14371443. (https://doi.org/10.1056/NEJM199511303332201)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Liu H, He J, Bagheri-Yarmand R, Li Z, Liu R, Wang Z, Bach DH, Huang YH, Lin P, Guise TA, et al.2022 Osteocyte CIITA aggravates osteolytic bone lesions in myeloma. Nature Communications 13 3684. (https://doi.org/10.1038/s41467-022-31356-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Loots GG, Kneissel M, Keller H, Baptist M, Chang J, Collette NM, Ovcharenko D, Plajzer-Frick I & & Rubin EM 2005 Genomic deletion of a long-range bone enhancer misregulates sclerostin in Van Buchem disease. Genome Research 15 928935. (https://doi.org/10.1101/gr.3437105)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lu J, Ji ML, Zhang XJ, Shi PL, Wu H, Wang C & & Im HJ 2017 MicroRNA-218-5p as a potential target for the treatment of human osteoarthritis. Molecular Therapy 25 26762688. (https://doi.org/10.1016/j.ymthe.2017.08.009)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Luis TC, Naber BAE, Roozen PPC, Brugman MH, De Haas EFE, Ghazvini M, Fibbe WE, Van Dongen JJM, Fodde R & & Staal FJT 2011 Canonical Wnt signaling regulates hematopoiesis in a dosage-dependent fashion. Cell Stem Cell 9 345356. (https://doi.org/10.1016/j.stem.2011.07.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lv F, Cai X, Yang W, Gao L, Chen L, Wu J & & Ji L 2020 Denosumab or Romosozumab therapy and risk of cardiovascular events in patients with primary osteoporosis: systematic review and meta- analysis. Bone 130 115121. (https://doi.org/10.1016/j.bone.2019.115121)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ma B, Landman EB, Miclea RL, Wit JM, Robanus-Maandag EC, Post JN & & Karperien M 2013 WNT signaling and cartilage: of mice and men. Calcified Tissue International 92 399411 22. (https://doi.org/10.1007/s00223-012-9675-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ma L, Zhao X, Liu Y, Wu J, Yang X & & Jin Q 2021 Dihydroartemisinin attenuates osteoarthritis by inhibiting abnormal bone remodeling and angiogenesis in subchondral bone. International Journal of Molecular Medicine 47 22. (https://doi.org/10.3892/ijmm.2021.4855)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mabille C, Ruyssen-Witrand A, Degboe Y, Gennero I, Loiseau HA, Roussel M, Hebraud B, Nigon D, Attal M & & Laroche M 2018 DKK1 and sclerostin are early markers of relapse in multiple myeloma. Bone 113 114117. (https://doi.org/10.1016/j.bone.2017.10.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mace ML, Gravesen E, Nordholm A, Egstrand S, Morevati M, Nielsen C, Kjaer A, Behets G, D'haese P, Olgaard K, et al.2021 Chronic kidney disease-induced vascular calcification impairs bone metabolism. Journal of Bone and Mineral Research 36 510522. (https://doi.org/10.1002/jbmr.4203)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mace ML, Gravesen E, Nordholm A, Egstrand S, Morevati M, Olgaard K & & Lewin E 2022 The calcified vasculature in chronic kidney disease secretes factors that inhibit bone mineralization. JBMR Plus 6 e10610. (https://doi.org/10.1002/jbm4.10610)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Magarò MS, Bertacchini J, Florio F, Zavatti M, Potì F, Cavani F, Amore E, De Santis I, Bevilacqua A, Reggiani Bonetti L, et al.2021 Identification of sclerostin as a putative new myokine involved in the muscle-to-bone crosstalk. Biomedicines 9 71. (https://doi.org/10.3390/biomedicines9010071)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mantovani A, Sani E, Fassio A, Colecchia A, Viapiana O, Gatti D, Idolazzi L, Rossini M, Salvagno G, Lippi G, et al.2019 Association between non-alcoholic fatty liver disease and bone turnover biomarkers in post-menopausal women with type 2 diabetes. Diabetes and Metabolism 45 347355. (https://doi.org/10.1016/j.diabet.2018.10.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Marchand A, Atassi F, Gaaya A, Leprince P, Le Feuvre C, Soubrier F, Lompre AM & & Nadaud S 2011 The Wnt/beta-catenin pathway is activated during advanced arterial aging in humans. Aging Cell 10 220232. (https://doi.org/10.1111/j.1474-9726.2010.00661.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Marenzana M, Greenslade K, Eddleston A, Okoye R, Marshall D, Moore A & & Robinson MK 2011 Sclerostin antibody treatment enhances bone strength but does not prevent growth retardation in young mice treated with dexamethasone. Arthritis and Rheumatism 63 23852395. (https://doi.org/10.1002/art.30385)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martín González C, Fernández Rodríguez CM, Abreu González P, García Rodríguez A, Alvisa Negrín JC, Cabañas Perales E, González Navarrete L, Vera Delgado VE, Ortega Toledo P & & González Reimers E 2022 Sclerostin in excessive drinkers: relationships with liver function and body composition. Nutrients 14 2574. (https://doi.org/10.3390/nu14132574)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Matsuoka K, Bakiri L, Wolff LI, Linder M, Mikels-Vigdal A, Patiño-García A, Lecanda F, Hartmann C, Sibilia M & & Wagner EF 2020 Wnt signaling and Loxl2 promote aggressive osteosarcoma. Cell Research 30 885901. (https://doi.org/10.1038/s41422-020-0370-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mcclung MR, Brown JP, Diez-Perez A, Resch H, Caminis J, Meisner P, Bolognese MA, Goemaere S, Bone HG, Zanchetta JR, et al.2018 Effects of 24 months of treatment with Romosozumab followed by 12 months of denosumab or placebo in postmenopausal women with low bone mineral density: a randomized, double-blind, Phase 2, parallel group study. Journal of Bone and Mineral Research 33 13971406. (https://doi.org/10.1002/jbmr.3452)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mcclung MR, Lewiecki EM, Cohen SB, Bolognese MA, Woodson GC, Moffett AH, Peacock M, Miller PD, Lederman SN, Chesnut CH, et al.2006 Denosumab in postmenopausal women with low bone mineral density. New England Journal of Medicine 354 821831. (https://doi.org/10.1056/NEJMoa044459)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McDonald MM, Reagan MR, Youlten SE, Mohanty ST, Seckinger A, Terry RL, Pettitt JA, Simic MK, Cheng TL, Morse A, et al.2017 Inhibiting the osteocyte-specific protein sclerostin increases bone mass and fracture resistance in multiple myeloma. Blood 129 34523464. (https://doi.org/10.1182/blood-2017-03-773341)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Miyatake K, Kumagai K, Imai S, Yamaguchi Y & & Inaba Y 2020 Sclerostin inhibits interleukin-1β-induced late stage chondrogenic differentiation through downregulation of Wnt/β-catenin signaling pathway. PLoS One 15 e0239651. (https://doi.org/10.1371/journal.pone.0239651)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Morales-Santana S, Garcia-Fontana B, Garcia-Martin A, Rozas-Moreno P, Garcia-Salcedo JA, Reyes-Garcia R & & Munoz-Torres M 2013 Atherosclerotic disease in type 2 diabetes is associated with an increase in sclerostin levels. Diabetes Care 36 16671674. (https://doi.org/10.2337/dc12-1691)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nakashima T, Hayashi M, Fukunaga T, Kurata K, Oh-Hora M, Feng JQ, Bonewald LF, Kodama T, Wutz A, Wagner EF, et al.2011 Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nature Medicine 17 12311234. (https://doi.org/10.1038/nm.2452)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Napoli N, Strollo R, Defeudis G, Leto G, Moretti C, Zampetti S, D'onofrio L, Campagna G, Palermo A, Greto V, et al.2018 Serum sclerostin and bone turnover in latent autoimmune diabetes in adults. Journal of Clinical Endocrinology and Metabolism 103 19211928. (https://doi.org/10.1210/jc.2017-02274)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nioi P, Taylor S, Hu R, Pacheco E, He YD, Hamadeh H, Paszty C, Pyrah I, Ominsky MS & & Boyce RW 2015 Transcriptional profiling of laser capture microdissected subpopulations of the osteoblast lineage provides insight into the early response to sclerostin antibody in rats. Journal of Bone and Mineral Research 30 14571467. (https://doi.org/10.1002/jbmr.2482)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Niziolek PJ, Bullock W, Warman ML & & Robling AG 2015 Missense mutations in LRP5 associated with high bone mass protect the mouse skeleton from disuse- and ovariectomy-induced osteopenia. PLoS One 10 e0140775. (https://doi.org/10.1371/journal.pone.0140775)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Nusse R & & Clevers H 2017 Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell 169 985999. (https://doi.org/10.1016/j.cell.2017.05.016)

  • Paton‐Hough J, Tazzyman S, Evans H, Lath D, Down JM, Green AC, Snowden JA, Chantry AD & & Lawson MA 2019 Preventing and repairing myeloma bone disease by combining conventional antiresorptive treatment with a bone anabolic agent in murine models. Journal of Bone and Mineral Research 34 783796. (https://doi.org/10.1002/jbmr.3606)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pelletier S, Confavreux CB, Haesebaert J, Guebre-Egziabher F, Bacchetta J, Carlier MC, Chardon L, Laville M, Chapurlat R, London GM, et al.2015 Serum sclerostin: the missing link in the bone-vessel cross-talk in hemodialysis patients? Osteoporosis International 26 21652174. (https://doi.org/10.1007/s00198-015-3127-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pols HAP, Felsenberg D, Hanley DA, Štepán J, Muñoz-Torres M, Wilkin TJ, Qin-Sheng G, Galich AM, Vandormael K, Yates AJ, et al.1999 Multinational, placebo-controlled, randomized trial of the effects of alendronate on bone density and fracture risk in postmenopausal women with low bone mass: results of the FOSIT study. Fosamax International Trial Study Group. Osteoporosis International 9 461468. (https://doi.org/10.1007/pl00004171)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Poole KES, Van Bezooijen RL, Loveridge N, Hamersma H, Papapoulos SE, Löwik CW & & Reeve J 2005 Sclerostin is a delayed secreted product of osteocytes that inhibits bone formation. FASEB Journal 19 18421844. (https://doi.org/10.1096/fj.05-4221fje)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Recker RR, Benson CT, Matsumoto T, Bolognese MA, Robins DA, Alam J, Chiang AY, Hu L, Krege JH, Sowa H, et al.2015 A randomized, double-blind phase 2 clinical trial of Blosozumab, a sclerostin antibody, in postmenopausal women with low bone mineral density. Journal of Bone and Mineral Research 30 216224. (https://doi.org/10.1002/jbmr.2351)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Regard JB, Cherman N, Palmer D, Kuznetsov SA, Celi FS, Guettier JM, Chen M, Bhattacharyya N, Wess J, Coughlin SR, et al.2011 Wnt/β-catenin signaling is differentially regulated by Gα proteins and contributes to fibrous dysplasia. PNAS 108 2010120106. (https://doi.org/10.1073/pnas.1114656108)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rodda SJ & & McMahon AP 2006 Distinct roles for Hedgehog and canonical Wnt signaling in specification, differentiation and maintenance of osteoblast progenitors. Development 133 32313244. (https://doi.org/10.1242/dev.02480)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Saag KG, Petersen J, Brandi ML, Karaplis AC, Lorentzon M, Thomas T, Maddox J, Fan M, Meisner PD & & Grauer A 2017 Romosozumab or alendronate for fracture prevention in women with osteoporosis. New England Journal of Medicine 377 14171427. (https://doi.org/10.1056/NEJMoa1708322)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sabbagh Y, Graciolli FG, O'brien S, Tang W, Dos Reis LM, Ryan S, Phillips L, Boulanger J, Song W, Bracken C, et al.2012 Repression of osteocyte Wnt/beta-catenin signaling is an early event in the progression of renal osteodystrophy. Journal of Bone and Mineral Research 27 17571772. (https://doi.org/10.1002/jbmr.1630)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sato AY, Cregor M, Delgado-Calle J, Condon KW, Allen MR, Peacock M, Plotkin LI & & Bellido T 2016 Protection from glucocorticoid-induced osteoporosis by anti-catabolic signaling in the absence of Sost/sclerostin. Journal of Bone and Mineral Research 31 17911802. (https://doi.org/10.1002/jbmr.2869)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Schenk R, Eggli P, Fleisch H & & Rosini S 1986 Quantitative morphometric evaluation of the inhibitory activity of new aminobisphosphonates on bone resorption in the rat. Calcified Tissue International 38 342349. (https://doi.org/10.1007/BF02555748)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Seefried L, Baumann J, Hemsley S, Hofmann C, Kunstmann E, Kiese B, Huang Y, Chivers S, Valentin MA, Borah B, et al.2017 Efficacy of anti-sclerostin monoclonal antibody BPS804 in adult patients with hypophosphatasia. Journal of Clinical Investigation 127 21482158. (https://doi.org/10.1172/JCI83731)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Semënov MV, Tamai K, Brott BK, Kühl M, Sokol S & & He X 2001 Head inducer Dickkopf-1 is a ligand for Wnt coreceptor LRP6. Current Biology 11 951961. (https://doi.org/10.1016/s0960-9822(0100290-1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Lüthy R, Nguyen HQ, Wooden S, Bennett L, Boone T, et al.1997 Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 89 309319. (https://doi.org/10.1016/s0092-8674(0080209-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Stolina M, Dwyer D, Niu QT, Villasenor KS, Kurimoto P, Grisanti M, Han CY, Liu M, Li X, Ominsky MS, et al.2014 Temporal changes in systemic and local expression of bone turnover markers during six months of sclerostin antibody administration to ovariectomized rats. Bone 67 305313. (https://doi.org/10.1016/j.bone.2014.07.031)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Storm T, Thamsborg G, Steiniche T, Genant HK & & Sorensen OH 1990 Effect of intermittent cyclical etidronate therapy on bone mass and fracture rate in women with postmenopausal osteoporosis. New England Journal of Medicine 322 12651271. (https://doi.org/10.1056/NEJM199005033221803)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sun M, Chen Z, Wu X, Yu Y, Wang L, Lu A, Zhang G & & Li F 2021 The roles of sclerostin in immune system and the applications of aptamers in immune-related research. Frontiers in Immunology 12 602330. (https://doi.org/10.3389/fimmu.2021.602330)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tagliaferri C, Wittrant Y, Davicco MJ, Walrand S & & Coxam V 2015 Muscle and bone, two interconnected tissues. Ageing Research Reviews 21 5570. (https://doi.org/10.1016/j.arr.2015.03.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Taylor S, Ominsky MS, Hu R, Pacheco E, He YD, Brown DL, Aguirre JI, Wronski TJ, Buntich S, Afshari CA, et al.2016 Time-dependent cellular and transcriptional changes in the osteoblast lineage associated with sclerostin antibody treatment in ovariectomized rats. Bone 84 148159. (https://doi.org/10.1016/j.bone.2015.12.013)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tian E, Zhan F, Walker R, Rasmussen E, Ma Y, Barlogie B & & Shaughnessy JD 2003 The role of the Wnt-signaling antagonist DKK1 in the development of osteolytic lesions in multiple myeloma. New England Journal of Medicine 349 24832494. (https://doi.org/10.1056/NEJMoa030847)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Toscani D, Bolzoni M, Ferretti M, Palumbo C & & Giuliani N 2018 Role of osteocytes in myeloma bone disease: anti-sclerostin antibody as new therapeutic strategy. Frontiers in Immunology 9 2467. (https://doi.org/10.3389/fimmu.2018.02467)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Turk JR, Deaton AM, Yin J, Stolina M, Felx M, Boyd G, Bienvenu JG, Varela A, Guillot M, Holdsworth G, et al.2020 Nonclinical cardiovascular safety evaluation of Romosozumab, an inhibitor of sclerostin for the treatment of osteoporosis in postmenopausal women at high risk of fracture. Regulatory Toxicology and Pharmacology 115 104697. (https://doi.org/10.1016/j.yrtph.2020.104697)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ueland T, Stilgren L & & Bollerslev J 2019 Bone matrix levels of Dickkopf and sclerostin are positively correlated with bone mass and strength in postmenopausal osteoporosis. International Journal of Molecular Sciences 20 2896. (https://doi.org/10.3390/ijms20122896)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Van Wesenbeeck L, Cleiren E, Gram J, Beals RK, Bénichou O, Scopelliti D, Key L, Renton T, Bartels C, Gong Y, et al.2003 Six novel missense mutations in the LDL receptor-related Protein 5 (LRP5) gene in different conditions with an increased bone density. American Journal of Human Genetics 72 763771. (https://doi.org/10.1086/368277)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vasiliadis ES, Evangelopoulos DS, Kaspiris A, Vlachos C & & Pneumaticos SG 2021 Sclerostin and its involvement in the pathogenesis of idiopathic scoliosis. Journal of Clinical Medicine 10 5286. (https://doi.org/10.3390/jcm10225286)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wijenayaka AR, Kogawa M, Lim HP, Bonewald LF, Findlay DM & & Atkins GJ 2011 Sclerostin stimulates osteocyte support of osteoclast activity by a RANKL-dependent pathway. PLoS One 6 e25900. (https://doi.org/10.1371/journal.pone.0025900)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Winkler DG, Sutherland MK, Geoghegan JC, Yu C, Hayes T, Skonier JE, Shpektor D, Jonas M, Kovacevich BR, Staehling-Hampton K, et al.2003 Osteocyte control of bone formation via sclerostin, a novel BMP antagonist. EMBO Journal 22 62676276. (https://doi.org/10.1093/emboj/cdg599)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Witcher PC, Miner SE, Horan DJ, Bullock WA, Lim KE, Kang KS, Adaniya AL, Ross RD, Loots GG & & Robling AG 2018 Sclerostin neutralization unleashes the osteoanabolic effects of Dkk1 inhibition. JCI Insight 3 e98673. (https://doi.org/10.1172/jci.insight.98673)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wu L, Guo H, Sun K, Zhao X, Ma T & & Jin Q 2016 Sclerostin expression in the subchondral bone of patients with knee osteoarthritis. International Journal of Molecular Medicine 38 13951402. (https://doi.org/10.3892/ijmm.2016.2741)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yamaguchi Y, Kumagai K, Imai S, Miyatake K & & Saito T 2018 Sclerostin is upregulated in the early stage of chondrogenic differentiation, but not required in endochondral ossification in vitro. PLoS One 13 e0201839. (https://doi.org/10.1371/journal.pone.0201839)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yao W, Dai W, Jiang L, Lay EYA, Zhong Z, Ritchie RO, Li X, Ke H & & Lane NE 2016 Sclerostin-antibody treatment of glucocorticoid-induced osteoporosis maintained bone mass and strength. Osteoporosis International 27 283294. (https://doi.org/10.1007/s00198-015-3308-6)

    • PubMed