SCLEROSTIN – a biomarker for predicting the onset of frailty
SCLEROSTIN, a protein that is predominantly produced by osteocytes, has gained considerable attention for its role in inhibiting bone formation (1). In addition to its effects on bone, sclerostin has been shown to have hormonal functions in non-skeletal tissues like adipocytes, blood vessels, muscles, and kidneys, where it plays a role in endothelial function, energy balance, glucose metabolism, physical performance, and kidney health (2-4). This broader systemic involvement highlights its potential effects on overall health.
Sclerostin circulates in the blood as a secreted protein and can easily be measured by ELISA assay technology, making it a promising biomarker for various age-related conditions, such as osteoporosis, sarcopenia, and cardiovascular diseases. The link between serum Sclerostin levels and frailty has not yet been studied. In a recent clinical study the relationship between circulating sclerostin levels and frailty, using both the phenotypic frailty model and the frailty index in a group of older adults has been investigated.
SCLEROSTIN – a biomarker for predicting the onset of frailty
“Methods: We collected blood samples from 244 older adults who underwent comprehensive geriatric assessments. Sclerostin levels were quantified using an enzyme-linked immunosorbent assay. Frailty was assessed using two validated approaches: the phenotypic model by Fried and the deficit accumulation frailty index (FI) by Rockwood.
Results: After controlling for sex, age, and body mass index, we found that serum sclerostin levels were significantly elevated in frail individuals compared to their robust counterparts (P<0.001). There was a positive correlation between serum sclerostin concentrations and the FI (P<0.001). Each standard deviation increase in serum sclerostin was associated with an odds ratio of 1.87 for frailty (P=0.003). Moreover, participants in the highest quartile of sclerostin levels had a significantly higher FI and a 9.91-fold increased odds of frailty compared to those in the lowest quartile (P=0.003 and P=0.039, respectively).
Conclusion: These findings, which for the first time explore the association between circulating sclerostin levels and frailty, have significant clinical implications, positioning sclerostin as one of potential blood-based biomarkers for frailty that captures the comprehensive physical, mental, and social aspects of the elderly, extending beyond its traditional role in bone metabolism.”
Sclerostin: From Molecule to Clinical Biomarker. Omran A, Atanasova D, Landgren F, Magnusson P. Int J Mol Sci. 2022 Apr 26;23(9):4751. doi: 10.3390/ijms23094751. PMID: 35563144; PMCID: PMC9104784.
Role of Sclerostin in Cardiovascular Disease. Golledge J, Thanigaimani S. Arterioscler Thromb Vasc Biol. 2022 Jul;42(7):e187-e202. doi: 10.1161/ATVBAHA.122.317635. Epub 2022 May 12. PMID: 35546488.
Sclerostin and Periostin associated with vascular risk scales in type 2 diabetes
Exciting research news! The BIOMEDICA bioactive Sclerostin and Periostin ELISA kits were used in a groundbreaking study evaluating the association of these bone proteins related to cardiovascular disease (CVD), with the main vascular risk scales in patients with type 2 diabetes.
All Biomedica ELISA assays are fully validated following international quality guidelines.
Sclerostin and Periostin associated with vascular risk scales in type 2 diabetes
Type 2 diabetes is linked to an elevated risk of cardiovascular disease (CVD), affecting approximately 35% of patients with the condition (1). As a result, assessing cardiovascular risk is essential for effective disease management in individuals with type 2 diabetes. Various risk scores have been developed to estimate CVD in the general population, including the Framingham Risk Score (FRS), the REGICOR and more recently, the SCORE2-Diabetes was introduced, specifically tailored for individuals with type 2 diabetes (2).
While these computational tools are utilized in clinical practice, there remains a need to investigate new biomarkers that could enhance cardiovascular risk stratification for patients with type 2 diabetes.
Typical bone proteins, including Sclerostin and Periostin, have been linked to cardiovascular disease (CVD). Concurrently, various risk scores have been created to forecast CVD in the general population. The objective of the following study was to examine the relationship between these bone proteins connected to CVD and key vascular risk scales:
Sclerostin and Periostin are associated with vascular risk in the SCORE2-Diabetes algorithm.
This suggests that Sclerostin and Periostin may serve as useful diagnostic biomarkers for vascular risk in patients with type 2 diabetes.
Future prospective studies are needed to validate the significance of these bone proteins in assessing vascular risk in the diabetic population.
Abstract
Background: Typical bone proteins, such as sclerostin and periostin, have been associated with cardiovascular disease (CVD). Simultaneously, several risk scores have been developed to predict CVD in the general population. Therefore, we aimed to evaluate the association of these bone proteins related to CVD, with the main vascular risk scales: Framingham Risk Score (FRS), REGICOR and SCORE2-Diabetes, in patients with type 2 diabetes. We focus in particular on the SCORE2-Diabetes algorithm, which predicts 10-year CVD risk and is specific to the study population.
Methods: This was a cross-sectional study including 104 patients with type 2 diabetes (62 ± 6 years, 60% males). Clinical data, biochemical measurements, and serum bioactive sclerostin and periostin levels were collected, and different risk scales were calculated. The association between bioactive sclerostin or periostin with the risk scales was analyzed.
Results: A positive correlation was observed between circulating levels of bioactive sclerostin (p < 0.001) and periostin (p < 0.001) with SCORE2-Diabetes values. However, no correlation was found with FRS or REGICOR scales. Both serum bioactive sclerostin and periostin levels were significantly elevated in patients at high-very high risk of CVD (score ≥ 10%) than in the low-moderate risk group (score < 10%) (p < 0.001 for both). Moreover, analyzing these proteins to identify patients with type 2 diabetes at high-very high vascular risk using ROC curves, we observed significant AUC values for bioactive sclerostin (AUC = 0.696; p = 0.001), periostin (AUC = 0.749; p < 0.001), and the model combining both (AUC = 0.795; p < 0.001). For diagnosing high-very high vascular risk, serum bioactive sclerostin levels > 131 pmol/L showed 51.6% sensitivity and 78.6% specificity. Similarly, serum periostin levels > 1144 pmol/L had 64.5% sensitivity and 76.2% specificity.
Conclusions: Sclerostin and periostin are associated with vascular risk in the SCORE2-Diabetes algorithm, opening a new line of investigation to identify novel biomarkers of cardiovascular risk in the type 2 diabetes population.
November is Diabetes Awareness Month bringing attention to diabetes and its impact on millions of individuals.
Diabetes mellitus has emerged as the third most significant non-communicable disease, following cardiovascular diseases and cancer. This condition encompasses a group of metabolic disorders marked by chronic hyperglycemia resulting from various causes, along with inadequate insulin secretion and impaired insulin action. According to the most recent statistics from the International Diabetes Federation, the global number of individuals with diabetes reached 530 million in 2021, with projections suggesting it could exceed 780 million by 2045. Due to the long-term nature of the disease, diabetes can lead to damage across multiple body systems or organs, resulting in various complications (1, 2). Beyond the more commonly known complications of diabetes such as heart disease, diabetes can also affect the skeletal system. This severe complication of diabetes leads to bone loss potentially resulting in osteoporosis and increased fracture risk.
Identifying biomarkers that may predict fracture risk in individuals with diabetes is crucial for improving patient care.
Sclerostin and Fracture Risk Prediction in Diabetes
Sclerostin (SOST) is a bone-related protein that is mainly produced by osteocytes, bone cells embedded in the bone matrix. Sclerostin is considered to be one of the major regulators of bone formation. It is a soluble antagonist of the Wnt signaling pathway and its inactivation leads to bone degradation, while the of Wnt signaling promotes bone formation (3). Sclerostin has been a target of therapeutic antibodies for osteoporosis treatment due to its role in inhibiting bone formation.
Bone as an endocrine organ
Research indicates that bone, which is involved in lipid and glucose metabolism, is increasingly recognized as an endocrine organ. Recent findings suggest that sclerostin contributes to disorders related to lipid and glucose metabolism (4). Studies have shown that Sclerostin levels are increased in individuals with prediabetes and correlated with insulin resistance in the skeletal muscle, liver, and adipose tissue (5). In addition, Sclerostin levels have been shown to be negatively associated with insulin sensitivity in obese but not in lead woman (6).
Further studies have revealed that increased serum Sclerostin levels are associated with vertebral fractures in patients with type 2 diabetes mellitus (7, 8). Sclerostin, has also been suggested to have predictive value for fracture risk in patients with diabetes (9).
Sclerostin – a promising circulating marker of diabetic bone disease
Sclerostin has emerged as a promising circulating marker of diabetic bone disease. It may not only reflect the degree of osteocyte dysfunction and the suppression of bone formation that occurs in this disease, but it may also potentially reflect the vascular alterations that are associated with specific bone alterations such as cortical porosity (10).
Additional research is essential to enhance the understanding of biochemical markers in the assessment of diabetic bone disease. Specifically, the ability of bone markers to forecast fracture risk needs further examination.
Circulating Sclerostin levels can reliably be measured in human serum and plasma samples with a conventional SCLEROSTIN ELISA Assay Kit.
The Biomedica SCLEROSTIN ELISA Assay Kit (# BI-20492) was utilized in a recent publication assessing the associations between serum and bone sclerostin levels and biomarkers of bone turnover and bone histomorphometry. Read more: Sclerostin, Osteocytes, and Wnt Signaling in Pediatric Renal Osteodystrophy.
Sclerostin a biomarker in renal pediatric bone disease
Sclerostin, Osteocytes, and Wnt Signaling in Pediatric Renal Osteodystrophy. Laster M. et al., Nutrients. 2023 Sep 25;15(19):4127. doi: 10.3390/nu15194127. PMID: 37836411; PMCID: PMC10574198 . link to full text
Abstract
The pathophysiology of chronic kidney disease-mineral and bone disorder (CKD-MBD) is not well understood. Specific factors secreted by osteocytes are elevated in the serum of adults and pediatric patients with CKD-MBD, including FGF-23 and sclerostin, a known inhibitor of the Wnt signaling pathway. The molecular mechanisms that promote bone disease during the progression of CKD are incompletely understood. In this study, we performed a cross-sectional analysis of 87 pediatric patients with pre-dialysis CKD and post-dialysis (CKD 5D). We assessed the associations between serum and bone sclerostin levels and biomarkers of bone turnover and bone histomorphometry. We report that serum sclerostin levels were elevated in both early and late CKD. Higher circulating and bone sclerostin levels were associated with histomorphometric parameters of bone turnover and mineralization. Immunofluorescence analyses of bone biopsies evaluated osteocyte staining of antibodies towards the canonical Wnt target, β-catenin, in the phosphorylated (inhibited) or unphosphorylated (active) forms. Bone sclerostin was found to be colocalized with phosphorylated β-catenin, which suggests that Wnt signaling was inhibited. In patients with low serum sclerostin levels, increased unphosphorylated “active” β-catenin staining was observed in osteocytes. These data provide new mechanistic insight into the pathogenesis of CKD-MBD and suggest that sclerostin may offer a potential biomarker or therapeutic target in pediatric renal osteodystrophy.
Related Literature
FGF-23 and sclerostin in serum and bone of CKD patients. Lima F, Monier-Faugere MC, Mawad H, David V, Malluche HH. Clin Nephrol. 2023 May;99(5):209-218. doi: 10.5414/CN111111. PMID: 36970967; PMCID: PMC10286735. (Biomedica Sclerostin ELISA Assay Kit, cat. no. BI-20492 citation)
Sclerostin and Dickkopf-1 in renal osteodystrophy. Cejka D, Herberth J, Branscum AJ, Fardo DW, Monier-Faugere MC, Diarra D, Haas M, Malluche HH. Clin J Am Soc Nephrol. 2011 Apr;6(4):877-82. doi: 10.2215/CJN.06550810. Epub 2010 Dec 16. PMID: 21164019; PMCID: PMC3069382. (Biomedica Sclerostin ELISA Assay Kit, cat. no. BI-20492 citation)
Type 2 diabetes mellitus (T2DM) is characterized by a persistent state of elevated blood sugar levels and glucose intolerance, resulting from the body´s incomplete response to insulin, accompanied by an increase in insulin production and a subsequent insulin deficiency. Individuals suffering from T2DM have an increased risk of cardiovascular disease (CVD). High glucose levels, insulin resistance, and chronic inflammation, contribute to endothelial dysfunction (ED) and atherosclerosis (1). ED refers to an impairment of the endothelium, the inner lining of blood vessels, which play an important role in regulating vascular health.
Sclerostin is associated with endothelial dysfunction in patients with type 2 diabetes
Sclerostin is a protein known primarily for its role in bone metabolism. It has also been identified of being linked to endothelial dysfunction in individuals diagnosed with type 2 diabetes (2). Sclerostin is predominantly secreted by osteocytes, cells that are embedded in the bone. However, vascular endothelial cells have also been observed to produce sclerostin leading to the discovery of its significant anti-calcifying role (3).
Sclerostin is associated with endothelial dysfunction in patients with type 2 diabetes: In an investigation in individuals with T2DM, researchers measured endothelial dysfunction by digital thermal monitoring (2). This method is a valid and noninvasive technique to evaluate endothelial function using temperature change on finger as a surrogate measure of the magnitude of vascular reactivity index (VRI) (4) . Serum Sclerostin levels were measured in the T2DM cohort with the Biomedica ELISA. The prospective cross-sectional study revealed that serum sclerostin levels are positively associated with endothelial dysfunction measured in patients with T2DM.
A previous cross-sectional study in patients with T2DM, with/without cardiovascular disease, determined Sclerostin levels and its expression by RT-qPCR and immunohistochemistry in calcified and non-calcified artery of the lower limb from T2D. Serum Sclerostin was measured with an ELISA from Biomedica. Moreover, in vitro experiments were performed in vascular smooth muscle cells under calcifying conditions investigating the cardioprotective function of Sclerostin (5). The study provided evidence that supports the protective function of Sclerostin in the development of vascular calcification. The findings suggest that Sclerostin could potentially reduce the susceptibility to atherosclerosis by decreasing atherosclerotic plaque formation and underscore the significance of the bone-vascular axis when developing therapeutic strategies for treating impaired bone metabolism or vascular diseases (5).
Features and Benefits when measuring Sclerostin with the Biomedica ELISA kits
November is “Diabetes Awareness Month” raising attention to this fast growing and life-threating epidemic. Patients suffering from diabetes have a risk of additional health complications, including heart disease, strokes, and diabetic kidney disease (DKD). People who develop DKD mostly have few symptoms in the early stage of the disease, although the risk of developing severe kidney damage is very high. High blood sugar levels may damage the small blood vessels in the kidney leading to kidney damage, kidney failure, and high blood pressure (1).
FGF23 and Sclerostin – novel biomarkers in diabetic kidney disease
Traditionally, the bone is regarded as a structural organ that gives the human body support and facilitates physical movement. However, bone is also a source of various hormones including fibroblast growth factor 23 and sclerostin that play an important role in regulating glucose metabolism and DKD (2).
FGF23 and Sclerostin – novel biomarkers in diabetic kidney disease
Fibroblast growth factor 23 (FGF23) is a bone-derived protein that regulates phosphate metabolism, by inhibiting renal phosphate reabsorption. There is increasing evidence that FGF23 plays a role in type 2 diabetes (T2DM), as FGF23 levels are elevated in these patients, even in individuals with preserved kidney function when compared to the general population (3). Phosphate independent effects on FGF23 following glucose loading were shown in a recent study demonstrating that FGF23 is associated with glucose, insulin and proinsulin levels, as well as obesity (4 ). Furthermore, FGF23 has also been shown to be associated with the development of gestational diabetes mellitus (5).
Sclerostin is a protein that is produced by bone cells that inhibits bone formation. Recent research suggests that Sclerostin also plays a role in lipid and glucose metabolism as serum sclerostin is negatively associated with insulin sensitivity as measured in obese, but not lean women (5). Sclerostin levels have also been shown to be increased in individuals with prediabetes (6).
FGF23 and Sclerostin can reliable by measured with conventional ELISA assays from BIOMEDICA.
Bone markers are currently used to monitor skeletal diseases and treatments. The proteins Sclerostin and Dickkopf-1 (DKK-1) reflect distinct biological processes and have gained attention as potential biomarkers for bone-related conditions. They may provide valuable information for diagnosis, prognosis, and monitoring of bone diseases and treatments.
Sclerostin and DKK-1 emerging biomarkers for bone disease
Sclerostin and Dickkkopf-1 are two important osteocyte proteins that are involved in the regulation of bone metabolism, particularly through their interactions with the Wnt signaling pathway.
SCLEROSTIN (SOST) is a glycoprotein that is primarily secreted by osteocytes, the most abundant cells in bone tissue. It inhibits Wnt signaling, which is a critical pathway regulating bone formation and remodeling. Sclerostin acts as a negative regulator of bone formation by binding to the LRP5/6 co-receptors (low-density lipoprotein receptor protein), which activate Wnt signaling. By binding to LRP5/6, sclerostin inhibits the interaction between Wnt ligands and the Frizzled receptor, thereby inhibiting Wnt signaling and suppressing bone formation. Inhibition of Sclerostin has led to the development of a novel anabolic therapy for osteoporosis.
DICKKOPF-1 (DKK-1) is a protein that also inhibits the Wnt signaling pathway. DKK-1 binds to the LRP5/6 co-receptors thereby preventing Wnt ligand interaction thus inhibiting bone formation and promoting bone resorption.
The Wnt-signaling pathway is one of the most important pathways controlling bone metabolism. Sclerostin and Dickkopf-1 act as Wnt inhibitors and play a crucial role in controlling bone formation and resorption.
Sclerostin and DKK-1 can easily be measured in blood samples with an ELISA assay
A healthy skeleton depends on a continuous renewal and maintenance of the bone tissue. The process of bone remodeling is highly controlled and consists of a fine-tuned balance between bone formation and bone resorption. Biochemical markers of bone turnover are already in use for monitoring diseases and treatment involving the skeletal system, but novel biomarkers reflecting specific biological processes in bone and interacting tissues may prove useful for diagnostic, prognostic, and monitoring purposes. The Wnt-signaling pathway is one of the most important pathways controlling bone metabolism and consequently the action of inhibitors of the pathway such as sclerostin and Dickkopf-related protein 1 (DKK1) have crucial roles in controlling bone formation and resorption. Thus, they might be potential markers for clinical use as they reflect a number of physiological and pathophysiological events in bone and in the cross-talk with other tissues in the human body. This review focuses on the clinical utility of measurements of circulating sclerostin and DKK1 levels based on preanalytical and analytical considerations and on evidence obtained from published clinical studies. While accumulating evidence points to clear associations with a number of disease states for the two markers, and thus, the potential for especially sclerostin as a biochemical marker that may be used clinically, the lack of standardization or harmonization of the assays still hampers the clinical utility of the markers.
Decrease of bone biomarker Sclerostin in women with anorexia nervosa during nutrition therapy – indication of reduced bone loss
Anorexia nervosa (AN) is an eating disorder and has one of the highest mortality rates of any mental illness. It affects roughly 2.9 million people and many experience bone loss and increased fracture risk. In a 3-year prospective study, Swedish researchers looked into the long-term effects of nutrition therapy. They investigated bone and mineral metabolism and biomarkers young women with AN. Their results showed that body mass index (BMI) and fat mass was increased. The regulatory bone biomarker Sclerostin decreased during nutrition therapy and further over 3 years, indicating reduced bone loss.
Svedlund A, Pettersson C, Tubic B, Ellegård L, Elfvin A, Magnusson P, Swolin-Eide D. J Bone Miner Metab. 2022 Aug 12. doi: 10.1007/s00774-022-01359-x. Epub ahead of print. PMID: 35960382.
Abstract
Introduction: Anorexia nervosa (AN) increases the risk of impaired bone health, low areal bone mineral density (aBMD), and subsequent fractures. This prospective study investigated the long-term effects of bone and mineral metabolism on bone and biomarkers in 22 women with AN.
Materials and methods: Body composition and aBMD were measured by dual-energy X-ray absorptiometry (DXA) and peripheral quantitative computed tomography. Total and free 25-hydroxyvitamin D (25OHD), C-terminal collagen cross-links (CTX), osteocalcin, bone-specific alkaline phosphatase (BALP), leptin, sclerostin, and oxidized/non-oxidized parathyroid hormone (PTH) were analyzed before and after 12 weeks of intensive nutrition therapy and again 3 years later. An age-matched comparison group of 17 healthy women was recruited for the 3-year follow-up.
Results: Body mass index (BMI) and fat mass increased from baseline to 3 years in women with AN. Sclerostin decreased during nutrition therapy and further over 3 years, indicating reduced bone loss. CTX was elevated at baseline and after 12 weeks but decreased over 3 years. BALP increased during nutrition therapy and stabilized over 3 years. Free 25OHD was stable during treatment but decreased over 3 years. Non-oxidized PTH was stable during treatment but increased over 3 years. Trabecular volumetric BMD in AN patients decreased during the first 12 weeks and over 3 years despite stable BMI and bone biomarkers implying increased BMD.
Conclusion: Our findings highlight the importance of early detection and organized long-term follow-up of bone health in young women with a history of AN.
Keywords: DXA; Eating disorder; Osteoporosis; Sclerostin; Vitamin D
Background: Little is known about the long-term outcome of anorexia nervosa.
Aims: To study the 30-year outcome of adolescent-onset anorexia nervosa.
Method: All 4291 individuals born in 1970 and attending eighth grade in 1985 in Gothenburg, Sweden were screened for anorexia nervosa. A total of 24 individuals (age cohort for anorexia nervosa) were pooled with 27 individuals with anorexia nervosa (identified through community screening) who were born in 1969 and 1971-1974. The 51 individuals with anorexia nervosa and 51 school- and gender-matched controls were followed prospectively and examined at mean ages of 16, 21, 24, 32 and 44. Psychiatric disorders, health-related quality of life and general outcome were assessed.
Results: At the 30-year follow-up 96% of participants agreed to participate. There was no mortality. Of the participants, 19% had an eating disorder diagnosis (6% anorexia nervosa, 2% binge-eating disorder, 11% other specified feeding or eating disorder); 38% had other psychiatric diagnoses; and 64% had full eating disorder symptom recovery, i.e. free of all eating disorder criteria for 6 consecutive months. During the elapsed 30 years, participants had an eating disorder for 10 years, on average, and 23% did not receive psychiatric treatment. Good outcome was predicted by later age at onset among individuals with adolescent-onset anorexia nervosa and premorbid perfectionism.
Conclusions: This long-term follow-up study reflects the course of adolescent-onset anorexia nervosa and has shown a favourable outcome regarding mortality and full symptom recovery. However, one in five had a chronic eating disorder.
Mitchell JE, Peterson CB. N Engl J Med. 2020 Apr 2;382(14):1343-1351. doi: 10.1056/NEJMcp1803175. PMID: 32242359.
Effect of Sclerostin Inhibition on Cardiovascular Safety for the Treatment of Severe Osteoporosis
Osteoporosis is a skeletal disorder characterized by diminished bone strength that is responsible for an increased fracture risk. The glycoprotein sclerostin acts as an inhibitor of bone formation. Therapies directed against this molecule have been developed. A humanized antibody against sclerostin has been approved for the treatment of severe osteoporosis in postmenopausal women in many parts of the world. A recent review by Langdahl BL and colleagues sumarizes the current knowledge of the effect of sclerostin inhibition on cardiovascular safety.
Bovijn J, Krebs K, Chen CY, Boxall R, Censin JC, Ferreira T, Pulit SL, Glastonbury CA, Laber S, Millwood IY, Lin K, Li L, Chen Z, Milani L, Smith GD, Walters RG, Mägi R, Neale BM, Lindgren CM, Holmes MV. Sci Transl Med. 2020 Jun 24;12(549):eaay6570. doi: 10.1126/scitranslmed.aay6570. PMID: 32581134; PMCID: PMC7116615.
Abstract
Inhibition of sclerostin is a therapeutic approach to lowering fracture risk in patients with osteoporosis. However, data from phase 3 randomized controlled trials (RCTs) of romosozumab, a first-in-class monoclonal antibody that inhibits sclerostin, suggest an imbalance of serious cardiovascular events, and regulatory agencies have issued marketing authorizations with warnings of cardiovascular disease. Here, we meta-analyze published and unpublished cardiovascular outcome trial data of romosozumab and investigate whether genetic variants that mimic therapeutic inhibition of sclerostin are associated with higher risk of cardiovascular disease. Meta-analysis of up to three RCTs indicated a probable higher risk of cardiovascular events with romosozumab. Scaled to the equivalent dose of romosozumab (210 milligrams per month; 0.09 grams per square centimeter of higher bone mineral density), the SOST genetic variants were associated with lower risk of fracture and osteoporosis (commensurate with the therapeutic effect of romosozumab) and with a higher risk of myocardial infarction and/or coronary revascularization and major adverse cardiovascular events. The same variants were also associated with increased risk of type 2 diabetes mellitus and higher systolic blood pressure and central adiposity. Together, our findings indicate that inhibition of sclerostin may elevate cardiovascular risk, warranting a rigorous evaluation of the cardiovascular safety of romosozumab and other sclerostin inhibitors.
The glycoprotein Sclerostin is mainly secreted by osteocytes and acts as a negative regulator of bone mass and strength by inhibiting bone formation. Studies have shown that high intensity exercise induces an increase in serum Sclerostin levels suggesting that it may be a key protein involved in muscle and bone interaction. A recent study by Śliwicka E and colleagues https://buff.ly/3qx6V4U evaluated the effects of a marathon race on selected myokines and Sclerostin in male recreational runners. Results show that in response to the marathon run, a complex network of endocrine interactions is initiated. Further research is needed to fully elucidate the long-term impact of prolonged high intensity exercise on the human body. Exercise-induced increase in sclerostin- related finding: https://buff.ly/3atiBA4
Planning your study? Contact us by e-mail or phone to receive your special study quote.
Serum levels of sclerostin reflect altered bone microarchitecture in patients with hepatic cirrhosis. Sclerostin, a glycoprotein secreted mainly by osteocytes, regulates bone mass by decreasing bone formation.
In patients with hepatic cirrhosis, areal bone mineral density (aBMD) is decreased especially at the lumbar spine. aBMD alone can be insufficient to explain increased fracture risk and bone microarchitecture can provide additional information. However, since assessment of bone microarchitecture is complex, biomarkers could help assess fracture risk. In a study of several biomarkers, Wakolbinger et al. found a correlation between sclerostin and altered bone microarchitecture in hepatic cirrhosis https://link.springer.com/article/10.1007/s00508-019-01595-8.
Biomedica´s bioactive sclerostin ELISA measures bioactive sclerostin by using a monoclonal antibody directed at the LRP5/6 binding region, capturing all circulating sclerostin forms containing the free-receptor binding site. It is validated in depth according to FDA quality standards, to ensure the ELISA reliability.
Researchers have identified the soluble WNT pathway inhibitor SCLEROSTIN as an independent risk factor for all-cause #mortality in patients after kidney transplantation. 600 stable renal transplant recipients were followed for all-cause mortality for 3 years.
Sclerostin is an independent risk factor for all-cause mortality in kidney transplant recipients. Zeng S et al., Clin Exp Nephrology (2020). Click link for full text.
√ HIGH QUALITY – fully validated assay according to ICH/FDA/EMEA guidelines √ LOW SAMPLE VOLUME – only 20 µl sample / well √ EASY – convenient ready to use protocol √ MOST REFERENCED Sclerostin ELISA
Also available: Bioactive Sclerostin ELISA https://www.bmgrp.com/product/cardiovascular/biomedica-bioactive-sclerostin-elisa-human-sost/ √ specific antibodies targeting the receptor binging region
The only Sclerostin ELISA that utilizes specific EPITOPE MAPPED ANTIBODIES enabling the analysis of bioactive Sclerostin in clinical samples.
HIGHLY SPECIFIC and DEFINED: capture antibody directed against Sclerostin’s bioactive site. Learn more
RELIABLE: human serum based calibrators and controls, rigorously validated
LOW SAMPLE VOLUME: 20 µl / well
QUICK: total incubation time 3.5 h
First bioactive Sclerostin ELISA for clinical samples
Areas of interest: osteoporosis, cancer induced bone diseases, rheumatoid arthritis, chronic inflammation, kidney diseases, therapy monitoring of anabolic treatment.
For detailed information please click corresponding links:
Background: Circulating serum sclerostin levels are supposed to give a good estimation of the levels of this negative regulator of bone mass within bone. Most studies evaluating total serum sclerostin found different levels in males compared to females and in older compared to younger subjects. Besides an ELISA detecting total sclerostin an ELISA determining bioactive sclerostin has been developed. The aim of this study was to investigate serum levels of bioactive sclerostin in an Austrian population-based cohort.
Methods: We conducted a cross-sectional observational study in 235 healthy subjects. Using the bioactive ELISA assay (Biomedica) bioactive sclerostin levels were evaluated.
Results: Serum levels of bioactive sclerostin were higher in men than in women (24%). The levels correlated positively with age (r = 0.47). A positive correlation could also be detected with body mass index and bone mineral density.
Conclusion: Using the ELISA detecting bioactive sclerostin our results are consistent with data in the literature obtained by different sclerostin assays. The determination of sclerostin concentrations in peripheral blood thus appears to be a robust parameter of bone metabolism.
Sclerostin ELISA | BI-20492 Highlights:
Rigorously validated according to FDA/ICH/EMEA guidelines
Specific antibodies targeting the receptor binding region
Rigorously validated for clinical samples according to FDA/ICH/EMEA guidelines
Low sample volume – 20 µl of serum/plasma per well
Biomedica’s FGF23 ELISA kits now available for the US Biotech community at 25% trial discount.
FGF23 (fibroblast growth factor 23) is a 32 kDa protein with 251 amino acids that is proteolytically processed between arginine179 and serine180 to generate N-terminal and C-terminal fragments. FGF23 is mainly secreted by osteocytes and controls phosphate and 1,25(OH)2 vitamin D homeostasis. Epidemiological data suggest that higher FGF23 concentrations are associated with all-cause mortality, cardiovascular mortality, a higher risk of myocardial infarction, stroke and heart failure. ➡️ Learn more:FGF23 – An Overview
Biomedica’s FGF23 ELISA kits, widely recognized in Europe and Asia, are now also available in the US.
💡 What makes them a game-changer?
✓ MULTI-MATRIX: for plasma, serum, cell-culture ✓ CONVENIENT: 50 µl sample/well, all buffers included ✓ RELIABLE: validated following quality guidelines ✓ COMPARABLE: good correlation with existing kits ✓ EASY HANDLING: 7 prediluted standards, 2 controls ✓ TRUSTED: cited in more than 80 publications
Biomedica offers world-leading quality products at competitive pricing.
FGF23 signalling and physiology. Ho BB, Bergwitz C. J Mol Endocrinol. 2021 Feb;66(2):R23-R32. doi: 10.1530/JME-20-0178. PMID: 33338030; PMCID: PMC8782161.
Kidney dysfunction causes disruptions in bone and mineral metabolism and changes in the regulators of the renal-bone axis through various mechanisms. Studies have demonstrated that fibroblast growth factor 23 (FGF23) and inflammatory markers rise, while iron status may decline (1, 2). In healthy individuals, FGF23 is regulated by phosphate, Vitamin D (1,25(OH)2D), and parathyroid hormone (PTH). However in the early stages of chronic kidney disease (CKD), FGF23 levels begin to increase even before plasma phosphate levels increase (3).
Thus, CKD causes disruptions in bone and mineral metabolism that also includes the regulatory hormones, resulting chronic kidney disease-mineral bone disorder (CKD-MBD).
Bone biomarkers in early renal impairment
A recent study investigated the effects of Vitamin D supplementation in a cohort of healthy community-dwelling older people. Baseline blood concentrations of bone regulatory markers including Sclerostin (SOST), Dickkopf-related Protein 1 (DKK1); Osteoprotegerin (OPG) and soluble RANKL (sRANKL), Fibroblast growth factor 23 (intact and c-terminal FGF23) and TNF-alpha, Interleukin-6 (IL-6) were analysed. The results showed that SOST, cFGF23, iFGF23, PTH and TNF-alpha were elevated in the group of individuals with early kidney impairment compared to those with normal kidney function. Following Vitamin D supplementation, only cFGF23, 25(OH)D, and IL-6 showed differences between the groups.
The study identified alterations in the renal bone-axis that occur prior to clinical monitoring of patients. Early diagnosis in the initial stages of renal impairment may offer opportunities for preventing the progression of renal disease and chronic kidney disease-mineral bone disorder (CKD-MBD).
Join us at the ASBMR (American Society of Bone and Mineral Research) Conference at booth number 145. The annual meeting is taking place in Toronto, ON, Canada from September 27-30, 2024.
The American Society of Bone and Mineral Research (ASBMR) meeting is considered to be the largest event worldwide covering fields in bone, mineral and musculoskeletal research. The meeting attracts over 2,500 participants across the globe, including both clinicians and researchers working in different disciplines and at all career levels. The meeting provides attendees with exciting opportunities to exchange knowledge and to learn about the latest scientific and medical advances in the field.
Discover some of our biomarker assays for clinical research in bone and mineral disorders
During the process of bone remodeling, bone cells release biomarkers that can aid in the evaluation of bone diseases and serve as valuable therapeutic targets. These bone biomarkers can be readily detected in serum and plasma samples using immunoassays.
A recent review by Nicolas H Hart et al., provides researchers and clinicians involved in bone and mineral metabolism with a comprehensive contemporary update on the Biological basis of bone strength: anatomy, physiology and measurement. J Musculoskelet Neuronal Interact. 2020; ;20(3):347-371.
Abstract
Understanding how bones are innately designed, robustly developed and delicately maintained through intricate anatomical features and physiological processes across the lifespan is vital to inform our assessment of normal bone health, and essential to aid our interpretation of adverse clinical outcomes affecting bone through primary or secondary causes. Accordingly this review serves to introduce new researchers and clinicians engaging with bone and mineral metabolism, and provide a contemporary update for established researchers or clinicians. Specifically, we describe the mechanical and non-mechanical functions of the skeleton; its multidimensional and hierarchical anatomy (macroscopic, microscopic, organic, inorganic, woven and lamellar features); its cellular and hormonal physiology (deterministic and homeostatic processes that govern and regulate bone); and processes of mechanotransduction, modelling, remodelling and degradation that underpin bone adaptation or maladaptation. In addition, we also explore commonly used methods for measuring bone metabolic activity or material features (imaging or biochemical markers) together with their limitations.
September is Blood Cancer Awareness Month raising awareness about the various types of blood cancers, including leukemia, lymphoma, myeloma, and other hematologic malignancies. The goal of this month-long initiative is to educate the public about the signs, symptoms, and the risks that are associated with these cancers, as well as the importance of early detection and treatment.
Some cancer treatments may lead to bone loss and increases the risk of osteoporosis and fractures. Learn more about the impact of intensive chemotherapy and glucocorticoid treatment on bone remodeling markers in children with acute lymphoblastic leukemia in the study described below (1).
The campaign also focuses on highlighting the latest advancements in blood cancer research and treatment, emphasizing that ongoing research is essential for improved outcomes and quality of life for patients.
September is Blood Cancer Awareness Month
Acute lymphoblastic leukemia (ALL) is a type of cancer that affects the blood and bone marrow. ALL is the most common cancer in children. Intensive chemotherapy has improved the 5-year survival rate for ALL patients up to 90%. However, this improved survival has resulted in long-term complications associated with chemotherapy, including negative effects on bone health including osteopenia/osteoporosis, osteonecrosis, and increased fragility fractures.
Skeletal health relies on a balance between bone resorption and bone formation
The health of the skeleton relies on a balance between bone resorption and bone formation, a process known as “bone remodeling,” which is regulated by the RANKL/RANK/osteoprotegerin (OPG) and Wnt/β-catenin signaling pathways.
Currently, there is a lack of data regarding the impact of intensive chemotherapy on bone remodeling markers in children with ALL. In a recent study, researchers in Italy investigated the role of bone remodeling markers in children with acute lymphoblastic leukemia after intensive chemotherapy and glucocorticoid (GC) treatment (1). The bone remodeling markers such as RANKL, OPG and the WNT signaling molecules Sclerostin and DKK-1 were also measured with ELISA kits from BIOMEDICA.
The researchers found “higher levels of RANKL and OPG in ALL children compared to the controls, with a balance of RANKL/OPG ratio, whereas the levels of sclerostin and DKK-1, the main inhibitors of osteoblastogenesis, were similar in the two groups of subjects. These results suggest that in our cohort of ALL subjects, the intensive chemotherapy and GCs increased osteoclastic activity and, thereby, bone resorption” (1).
About the RANK/RANKL/OPG system
The receptor-ligand duo, RANK and RANKL are crucial regulators of bone metabolism and osteoclast development. Osteoprotegerin (OPG) acts as a decoy receptor for RANKL. It competitively binds to RANKL and interferes with the interaction of RANKL–RANK, thus blocking bone resorption. Beyond its role as a regulator of bone remodeling, the RANK/RANKL/OPG system has direct effects on the development of tumor cells, as it is implicated in the progression of breast and prostate cancer as well as leukemia.
About the WNT signaling molecules Sclerostin and DKK-1
Sclerostin (SOST) is mainly produced by osteocytes and is considered as the major regulator of bone formation. It is a soluble antagonist of the Wnt signaling pathway. Inactivating this pathway leads to bone degradation, while induction of Wnt signaling promotes bone formation.
Dickkopf-1 (DKK-1) is an extracellular protein. DKK-1 plays a role in the regulation of bone metabolism, and is a secreted inhibitor of the Wnt signaling pathway. DKK-1 inhibits the differentiation of osteoblasts and thereby bone formation. The dysregulation of DKK1 can lead to cancer progression.
Biomedica offers quality kits for the measurement of these bone biomarkers
We are present in over 60 countries, ensuring global availability and customer support through partnerships with selected local distributors on almost every continent. Regardless of your location, we are here to help you reach your goals.
With over 30 years of experience, we specialize in developing and manufacturing high-quality ELISA assay kits for biomarkers in clinical research.
BIOMEDICA develops and manufactures high quality and widely cited immunoassays for clinical and pre-clinical applications in bone and cardiorenal diseases. For specific biomarkers, Biomedica has become a world-wide leader, e.g. Sclerostin, free sRANKL, OPG, DKK-1, and NT-proCNP.
Rath et al., Transl Lung Cancer Res; 2024; 13 (1), 5-15. PMID: 38405004
July is acknowledged as Bone Cancer and Sarcoma Awareness Month, focusing on increasing awareness about rare and challenging cancers affecting children and adolescents. Sarcoma is a type of cancer that originates in the bones and soft tissues. Soft tissues include muscle, fat, blood vessels, fibrous tissues such as tendons and ligament. More than 70 different subtypes of sarcomas have been reported. However, all sarcomas can be divided into two main groups: soft tissue sarcomas and bone cancers.
Soft Tissue Sarcomas is a rare type of cancer that originates from the growth of cells within the body´s soft tissue. More than 50 different soft tissue sarcomas have been reported (3 ) Soft tissue sarcoma can occur anywhere in the body, but it most frequently develops in arms, legs, and abdomen (4).
Osteosarcoma is the most prevalent type of bone cancer in children and adolescents with and incidence of around 4.4. cases per million children reported each year (1). Genomic alterations, particularly the inactivation of TP53 and RB, are present in most cases of osteosarcoma.
Ewing sarcoma is the second most common bone tumor occurring most frequently in teenagers, with a median age of 15 years. Ewing sarcomas is an aggressive tumor that develops usually in bone, but sometimes in soft tissue, most commonly affecting the lower extremity and pelvis. At diagnosis, up to 25% of patients , commonly found in the lung, bones, and bone marrow (2). This condition is biologically driven by a chromosomal translocation, typically involving the EWS and FLI1 genes.
July is Sarcoma and Bone Cancer Awareness Month
5 Things to Know About Bone Cancer and Sarcoma Awareness Month- link
Wnt Signalling molecules Sclerostin (SOST) and Dickkopf-1 (DKK-1) in Sarcoma
SCLEROSTIN has been shown to be expressed in bone and cartilage forming skeletal tumors. Sclerostin is widely localized to areas in osteoblastic differentiation and ossification (5). In a study using an osteosarcoma mouse model, the administration of sclerostin resulted in inhibited tumor growth and extended survival periods (6).
DKK-1 is a wnt inhibitor that has been shown to partially improve osteosarcoma survival by upregulating aldehyde-dehydrogenase-1A1, neutralizing reactive oxygen species originating from nutritional stress and chemotherapeutic challenge (7). In a mouse model researches demonstrated the use of a DKK-1 targeting vivo morpholino that reduces tumour progression (7).
FGF23 in uterine sarcoma
Human FGF23 has been shown to be highly expressed in uterine sarcoma highlighting its potential as a biomarker for the diagnosis and prognosis of the disease (8).
Sclerostin, DKK-1 and FGF23 can easily be measured in blood, urine and cell culture supernatants with an ELISA assay.
Sclerostin expression in skeletal sarcomas. Shen J, Meyers CA, Shrestha S, Singh A, LaChaud G, Nguyen V, Asatrian G, Federman N, Bernthal N, Eilber FC, Dry SM, Ting K, Soo C, James AW. Hum Pathol. 2016 Dec;58:24-34. doi: 10.1016/j.humpath.2016.07.016. Epub 2016 Aug 3. PMID: 27498059; PMCID: PMC6560186.
Antitumor Effect of Sclerostin against Osteosarcoma. Ideta H, Yoshida K, Okamoto M, Sasaki J, Kito M, Aoki K, Yoshimura Y, Suzuki S, Tanaka A, Takazawa A, Haniu H, Uemura T, Takizawa T, Sobajima A, Kamanaka T, Takahashi J, Kato H, Saito N. Cancers (Basel). 2021 Nov 29;13(23):6015. doi: 10.3390/cancers13236015. PMID: 34885123; PMCID: PMC8656567.
Join us at the International Conference on Children’s Bone Health
Click here for more information about the conference for anyone who is interested in
bone metabolism and bone mass in children, adolescents and young adults.
The ICCBH conference aims to unite researchers, clinicians, health professionals, and others from different fields to gain an understanding of the developing skeleton with regards to childhood health and disease. Latest advancements, innovative therapies, and genetic discoveries will be discussed.
More about Biomarkers in Bone Biology
Bone cells release biomarkers during bone remodeling. They can be used in assessing bone diseases and represent useful therapeutic targets. Bone biomarkers can easily be detected in serum and plasma samples by immunoassay.
Machine Learning for Bone Biomarker Profiling in Rheumatoid Arthritis
Rheumatoid arthritis (RA) is a chronic, progressive inflammatory disorder which can lead to severe joint damage and disability. In 2019, an estimated 18 million people worldwide were living with this disease (1). Untreated RA can lead to destruction of the joints as well as heart, lung or nervous system problems (2). Skeletal bone loss, referred to as osteopenia or osteoporosis, is a key feature of RA.
Sclerostin and Dickkopf-1 (DKK-1) are Wnt signaling proteins that are secreted by osteocytes, bone cells embedded in the bone matrix. They are inhibitors of bone formation and play a key role in the pathogeneses of systemic and localized bone loss in RA (3, 4). Serum levels of Sclerostin and DKK-1 have shown to be elevated in patients with RA compared to controls and correlate with bone erosions and inflammation (4, 5, 6). Findings in mice have demonstrated that DKK-1 triggers inflammatory bone degradation and neutralization of DKK-1 protects from systemic bone loss during inflammation (7). Interestingly, blocking the bone destruction molecule Sclerostin with an anti-sclerostin antibody has shown to be effective for the treatment of osteoporosis but may not be safe for patients suffering from inflammatory RA: in a rodent RA model, Weymeyer et al. demonstrated that Sclerostin inhibition did not stop bone loss and worsened clinical RA outcome by promoting TNF-dependent inflammatory joint destruction (8).
Controlling inflammation by biological therapies targeting pro-inflammatory cytokines has shown to have a positive effect in RA patients (5). Interleukin-6 is a key immunomodulatory cytokine that plays an important role in the development of RA. Inhibition of IL-6 has proven to be effective in treating patients with RA (9). A study by Briot et al showed that DKK-1 levels decreased in RA patients treated with an anti-IL-6 inhibitor (6).
Machine Learning for Bone Biomarker Profiling in Rheumatoid Arthritis
A recent cross-sectional study by Adami G et al., with over 1800 enrolled participants diagnosed with RA, Psoriatic Arthritis (PsA), and Systemic Sclerosis (SSc), employed machine learning techniques to assess the capability of biomarker profiles in differentiating RA patients from individuals with PsA and SSc. The Wnt signaling antagonists Sclerostin and Dickkopf-1 (DKK-1) were among the biomarkers measured. The study provided an in-depth understanding into the bone signature of RA that is marked by changes in bone mineral density and by unique biomarker profiles (6). Serum Sclerostin and DKK-1 levels were measured with ELISA assay kits from Biomedica.
GBD 2019: Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. https://vizhub.healthdata.org/gbd-results
Sclerostin inhibition promotes TNF-dependent inflammatory joint destruction. Wehmeyer C, Frank S, Beckmann D, Böttcher M, Cromme C, König U, Fennen M, Held A, Paruzel P, Hartmann C, Stratis A, Korb-Pap A, Kamradt T, Kramer I, van den Berg W, Kneissel M, Pap T, Dankbar B. Sci Transl Med. 2016 Mar 16;8(330):330ra35. doi: 10.1126/scitranslmed.aac4351. Epub 2016 Mar 16. PMID: 27089204.
Kidney disease is estimated to affect more than 800 million people worldwide (1). Taking action for prevention, early diagnosis and treatment can reduce the risk and the burden of kidney disease.
Role of the kidneys, kidney disease and risk factors
Kidneys are vital organs that regulate fluid balance, blood pressure and produce hormones that stimulate the production of red blood cells . Kidney disease is a condition in which kidneys lose their ability to effectively filter waste products and excess fluids from the blood. Kidney disease commonly leads to a decline in kidney function that may lead to kidney failure, characterized by the complete loss of kidney function. At this stage dialysis or kidney transplantation become the only treatment option.
Kidney problems can emerge suddenly (acute) or gradually over time (chronic). Various conditions, diseases and medications can contribute to acute and chronic kidney problems. Chronic kidney disease (CKD) is characterized by a prolonged period of kidney abnormalities that last for more than three months (2), whereas acute kidney disease – acute kidney injury (AKI) is characterized by a sudden loss of excretory kidney function (3).
Other forms of kidney disease include polycystic kidney disease (PKD) a genetic disorder that leads to kidney enlargement and impaired kidney function over time and glomerulonephritis (GN). GN is a group of diseases characterized by inflammation of the glomeruli, the filtration units of the kidney (4).
The most common risk factors for the development and progression of CKD are diabetes and high blood pressure. Managing blood sugar and blood pressure can help keep kidneys healthy. Other risk factors of CKD include heart disease, obesity, family history and genetic background as well as age, smoking and nutrition (5).
Consequences of kidney disease include heart disease, high blood pressure, bone and mineral disorders, and anemia (6).
Keep Your Kidneys Healthy
Regular exercise, weight control, a balanced diet (7) and sufficient fluid intake are only some of the ways to keep your kidney healthy.
World Kidney Day – about kidney health download here and check out the 6-Step Guide to Protecting Kidney Health here .
BIOMEDICA – Biomarker ELISA kits for clinical research in kidney disease
HIGH QUALITY ASSAYS – Fully validated according to international quality guidelines
Rare Disease Day is a global initiative held on the last day of February. It raises awareness for rare diseases to improve accessibility to medical treatment and representation for individuals diagnosed with rare diseases. It is estimated that around 300 million people worldwide are living with rare diseases.
Rare metabolic bone diseases are caused by genetic disorders that may directly or indirectly have an impact on bone structure or function (1). Other factors like hormones, tumors, diet or certain medications may also lead to abnormal growth and development of the skeleton. Some of the diseases are inherited many caused by genetic mutations. Other bone disorders are not inherited and can develop after birth.In some cases, the precise cause remains unknown.
Rare bone diseases
Rare bone diseases account for 5% of all birth defects and are an important cause of disability worldwide, yet they remain a difficult group of conditions to treat (2). It is estimated that more than 400 developmental abnormalties of the skeletal system exist (3).
The main rare metabolic bone diseases include Hypophosphatemia, Osteogenesis Imperfecta, Tumor-Induced Osteomalacia, X-Linked Hypophashatemia, and other Rare Bone Diseases (Fibrous Dysplasia, Osteopetrosis, High Bone Mass..) (4).
Biomarkers in Rare Bone Diseases
Biomarkers in Rare Bone Diseases provide a way to accelerate medical research by providing valuable insights into disease mechanisms. They play an important role in monitoring disease progression, optimizing treatments. and developing novel therapies.
NT-proCNP
C-natriuretic peptide (CNP) and its receptor, natriuretic peptide receptor-B (NPR-B) are important regulators of endochondral ossification and longitudinal bone growth (5, 6) The discovery and understanding of their physiological functions in promoting longitudinal bone growth have created opportunities for a specific targeted strategy in achondroplasia, the most common form of human dwarfism (7).
RANKL and OPG
Receptor activator of nuclear factor (NF-kappaβ) ligand (RANKL), its cellular receptor, receptor activator of NF-kappaβ (RANK), and the decoy receptor osteoprotegerin (OPG) are part of a cytokine system that regulate bone formation and resorption (8) . Denosumab is a bone anti-resorptive drug, a monoclonal antibody that binds RANKL and disrupts bone resorption. It has been approved for the treatment of osteoporosis and other bone-related disorders. The use of Denosumab in pediatric patients with Osteogenesis Imperfecta (OI), a genetic bone disorder, also known as brittle bone disease, shows decreased fractures and improved bone growth (9). A clinical trial at the National Institutes of Health found that Denosumab, significantly reduced abnormal bone turnover in adults with fibrous dysplasia, a rare disease characterized by weak, oddly shaped, or broke bones.
SCLEROSTIN
Sclerostin is a secreted protein that decreases bone formation. It binds to LRP-5 receptor on the surface of osteoblasts and consequently interferes with WNT signalling. Genetic sclerostin deficiency leads to increased bone formation and sclerotic bone disorders. Sclerostin inhibition is being evaluated as a potential approach to increase bone mass in Osteogenesis Imperfecta (10).
FGF23
Fibroblast growth factor 23 (FGF23) is a hormone that is produced by bone. It regulates serum phosphate levels by suppressing phosphate reabsorption in the kidney. Excessive actions of FGF23 are responsible for different kinds of hypophosphatemic rickets as found in X-linked hypophosphatemia (XLH) and osteomalacia. XLH is characterized by deformities of the lower limb and short stature. An anti-fibroblast growth factor-23 (FGF23) monoclonal antibody (Burosumab) has been approved as a novel treatment for hypophopshatemic rickets (11).
Measurement of FGF23 is a critical tool to assist in the evaluation and diagnosis of hypophosphatemic conditions (12, 13).
The second most common genetic form of hypophosphatemic rickets after XLH, is autosomal-dominant hypophosphatemic rickets (ADHR). ADHR is caused by specific mutations in the FGF23gene.
FGF23 can reliably be measured with an immunoassay (14).
Determination of FGF23 Levels for the Diagnosis of FGF23-Mediated Hypophosphatemia. Hartley IR, Gafni RI, Roszko KL, Brown SM, de Castro LF, Saikali A, Ferreira CR, Gahl WA, Pacak K, Blau JE, Boyce AM, Salusky IB, Collins MT, Florenzano P. J Bone Miner Res. 2022. 37(11):2174-2185. doi: 10.1002/jbmr.4702. PMID: 36093861; PMCID: PMC9712269.
The enzyme-linked immunosorbent assay (ELISA or EIA) is a laboratory method to detect and quantify the presence of a protein in biological samples (1, 2).
ELISA Assay Principle
When selecting an ELISA kit, researchers are often confronted with the question which assay to choose of the many commercially available kits.
It can be a challenge! Here are a few hints that may help.
HOW TO CHOOSE THE RIGHT ELISA KIT
As a general rule, before purchasing an assay, always read the protocol booklet (instructions for use – IFU or package insert) in detail. This should ensure that the kit will be suitable for your requirements. Check out the following:
1.ANALYTE
Which protein biomarker will you be measuring? Be sure to use the correct term during your search. Some biomarkers have alternative names (e.g. Sclerostin or SOST ELISA (SOST is actually the name for the gene that encodes Sclerostin).
2. SPECIES – SPECIFICITY – CROSS REACTIVITY
Verify if the assay can be used in the respective model such as e.g. human, rat, or mouse. Due to high homology between species, some ELISA kits work both in humans and in different animal species. As an example the biomarker ELISA kit for NT-proANP was developed for human use but due to the high sequence homology between species, the kit is successfully used to measure NT-proANP as a cardiac safety biomarker in various animal models (rat, mouse, rabbit, monkey).
3. SAMPLE TYPE
What is the sample type (matrix) you´ll be using (e.g. serum, EDTA-plasma, heparin-plasma, citrate-plasma, cell culture supernatants, urine..) ?
Verify if the assay is compatible for your sample type: check the package insert and, if available, check if there are validation data showing results (often found on the manufacturers website).
Of note: analysis of some biomarkers in the “wrong” matrix can lead to “false” results due to a matrix effect.
4. SAMPLE VOLUME
Check the amount of sample required per well (calculate volume to measure your samples in duplicates). Low sample volumes and precious samples are often a selection criterium.
5. SENSITIVTY – BIOMARKER CONCENTRATIONS TO BE EXPECTED
Before choosing an assay, look into the validation data of the kit (often documented in the ELISA protocol booklet or in the validation data files).
Reference values and pathological values in serum and/or plasma of the biomarker of interest are sometimes documented as well. These data can be helpful in selecting an appropriate assay. In some cases samples may require a pre-dilution. Therefore, always verify if the dilution buffer (assay buffer) is included in the kit or ask the assay developer for their input.
Of note: assays offering high sensitivity offer a different dynamic range than assays with a lower sensitivity. The dynamic range of an assay indicates the range of concentrations over which an assay can accurately quantify the analyte.
6. ASSAY PERFORMANCE – ASSAY VALIDATION
Careful evaluation of the assay´s performance characteristics is important in selecting an ELISA kit.
Choose an assay that has gone through a rigorous validation process. Check out if you can find data on the following performance characteristics:
Accuracy- detection of a protein biomarker in clinical samples.
Dilution linearity and parallelism – recovery of the analyte of interest in diluted samples
Specificity & cross-reactivity – making sure that you detect only the analyte of interest
Precision – within-run and in-between run precision – ensuring precise and reproducible results within an across assay lots
Calibration – ensures consistent performance over the range of the assay of the calibration curve
Sample stability – ensures the stability of the analyte of interest (e.g. exposure of real samples to multiple freeze-thaw cycles, stability at room temperature..).
ELISA Assay – microtiter plate
7. KIT COMPONENTS
Verify if the contents of the ELISA kit includes all the necessary components e.g. controls, assay dilution buffer. Consider storage requirements such as temperature sensitivity and expiration date.
8. CITATIONS & REFERENCES
Check if there are citations on the manufacturers website for the specific ELISA kit. Look into publications and seek feedback from researchers who have used the assay you are considering.
9. PRODUCT ORIGIN
Verify if the kit supplier is the kit manufacturer. More and more kits are repacked and are sold under different names, although it is always the same kit.
ELISA kit manufacturers will more likely give you qualified support as they “know” their product (e.g. availability of additional calibrators, controls, buffers.., technical know-how on the kit..).
10. CUSTOMER SUPPORT
Verify if the kit provider can provide timely and helpful customer service.
Duchenne muscular dystrophy (DMD) is a hereditary neuromuscular disease that leads to progressive muscle fiber degeneration and weakness. There is no cure for this disease and current therapy consists on treatment with glycocorticoids (GC). GC therapy is linked to risk of bone loss and increased fracture risk. Despite their adverse effects GC remain the standard care to slow down disease progression (2).
Steroid therapy and fracture prevention in Duchenne Muscular Dystrophy
This recent study explored factors that are associated with incident fracture risk in glucocorticoid (GC)-treated patients with Duchenne muscular dystrophy (DMD): Risk Factors Associated with Incident Vertebral Fractures in Steroid-treated Males with Duchenne Muscular Dystrophy. Brief, vertical fractures (VF) were prospectively evaluated in 38 males with Duchenne muscular dystrophy at study baseline and 12 months . The authors concluded the following: ” The observation that ≥ 1 prevalent VF and/or non-VF were the strongest predictors of incident VFs at 12 months supports the need for prevention of first fractures in this high-risk setting. Bone age delay, a marker of GC exposure, may assist in the prioritization of patients in efforts to prevent first fractures.”
Steroid therapy and fracture prevention in Duchenne Muscular Dystrophy – The Biomedica IL-6 and Sclerostin ELISA were highlighted in this study.
Purpose: Prevention of fractures is an unmet need in glucocorticoid (GC)-treated Duchenne muscular dystrophy. This study explored factors associated with incident vertebral fractures (VFs) to inform future fracture prevention efforts. Methods: VFs were evaluated prospectively at study baseline and 12 months on lateral spine radiographs in participants aged 4 to 25 years with Duchenne muscular dystrophy. Clinical factors were analyzed for their association with the change in Spinal Deformity Index (sum of the Genant-defined VF grades from T4 to L4) between baseline and 12 months. Results: Thirty-eight males were evaluated (mean ± SD age at baseline 11.0 ± 3.6 years; mean ± SD GC duration at baseline 4.1 ± 3.1 years; 74% ambulatory). Nine of 38 participants (24%) had 17 incident VFs, of which 3/17 VFs (18%) were moderate/severe. Participants with 12-month incident VF had lower mean ± SD baseline lumbar spine areal bone mineral density Z-scores (-2.9 ± 1.0 vs -1.9 ± 1.1; P = .049) and lower total body less head areal bone mineral density Z-scores (-3.1 ± 1.2 vs -1.6 ± 1.7; P = .036). Multivariable linear regression showed that at least 1 VF at baseline (P < .001), a higher number of antecedent non-VF (P < .001), and greater bone age delay at baseline (P = .027) were significant predictors of an increase in the Spinal Deformity Index from baseline to 12 months. Conclusion: The observation that ≥ 1 prevalent VF and/or non-VF were the strongest predictors of incident VFs at 12 months supports the need for prevention of first fractures in this high-risk setting. Bone age delay, a marker of GC exposure, may assist in the prioritization of patients in efforts to prevent first fractures.
2. Emerging therapies for Duchenne muscular dystrophy. Markati T, Oskoui M, Farrar MA, Duong T, Goemans N, Servais L. Lancet Neurol. 2022 Sep;21(9):814-829. doi: 10.1016/S1474-4422(22)00125-9. Epub 2022 Jul 15. PMID: 35850122.
Bone Health & Osteoporosis – Biomarkers of Bone Regulation
Maintaining strong and healthy bones is essential for our well-being. Bone not only provides structural support for the body but also protects vital organs and serves as a provider for minerals such as calcium and phosphorus. Bone density and bone strength are key components of bone health.
Osteoporosis is a condition of weakened and fragile bones. It is the most common metabolic bone disease in the world (1) that can affect individuals of various ages, but is more commonly associated with aging. Both men and women can be affected, but postmenopausal women are at higher risk to develop Osteoporosis due to the decline of hormonal estrogen levels which plays a protective role in bone. Prevention of Osteoporosis includes a balanced diet and exercise.
Bone remodeling is a continuous process that is tightly regulated between bone resorption of old or damaged bone and the formation of new bone. Various hormones and factors are involved in bone metabolism. The bone cycle consists of different phases and markers of bone metabolism can be categorized as markers of bone formation, markers of bone resorption and markers of the regulation of bone metabolism.
Bone Health & Osteoporosis – biomarkers of bone regulation
Protein biomarkers are often used in clinical research or clinical settings to assess bone health and to monitor the effectiveness of Osteoporosis treatments. Some of these biomarkers provide information about the regulatory processes involved in bone metabolism and turnover.
Monitoring these biomarkers can provide information on the overall health of bones. Some of these regulatory biomarkers include:
Sclerostin (SOST) – is produced by osteocytes, bone cells embedded in the bone. Sclerostin is a bone specific Wnt pathway inhibitor, that negatively regulates bone formation, by promoting osteoclastogenesis and bone resorption (3). Elevated Sclerostin levels may indicate decreased bone formation.
Dickkopf-1 (DKK-1) – is like Sclerostin an inhibitor of Wnt signaling, which is crucial for bone formation. Elevated serum DKK-1 promote bone resorption (4). DKK-1 levels may indicated suppressed bone formation.
Receptor Activator of Nuclear Factor-Kappa B Ligand (RANKL) – is a key regulator of osteoclast activation and formation. RANKL promotes bone resorption by activating the bone osteoclasts. RANKL is secreted by osteocytes and is the most important factor of osteoclast formation (5).
Osteoprotegerin (OPG) – is the decoy receptor for RANKL. OPG regulates bone resorption (6). Changes in the OPG / RANKL ratio can affect bone remodeling.
Fibroblast growth factor (FGF23) – is a hormone that regulates phosphate homeostasis and vitamin D metabolism. Abnormal FGF23 levels are associated with disorders affecting bone health e.g. hypophosphatemic rickets (7).
These protein biomarkers can easily be measured in human blood samples with an ELISA assay
With advancing age, the human body´s natural capacity to consistently renew bones and maintain their resilience diminishes. This decline is enhanced by conditions like osteoporosis. This poses a significant health concern for the elderly and is becoming a growing economic challenge on society. In an effort to address this problem, scientists are actively seeking novel therapeutic strategies to enhance bone regeneration.
Building bone with bioinspired molecules
By employing computer models and simulations, a Dresden-based team created innovative bio-inspired compounds that enhance bone regeneration in mice. These compounds can be incorporated into biomaterials, allowing for their localized introduction into bone defects. These newly developed molecules derive from glycosaminoglycans, which are extended sugar chains like hyaluronic acid or heparin. These molecules could be used to turn-off proteins like DKK-1 and Sclerostin that block bone regeneration that could lead to new and more effective therapies for bone diseases.
DKK-1 and SCLEROSTIN are two important proteins that play significant roles in regulating bone health and development.
DKK-1 (Dickkopf-1) acts as an inhibitor of the Wnt signaling pathway. This pathway is crucial for bone formation. Overexpression of DKK-1 leads to a decrease in bone formation characterized by conditions with impaired bone density and increased fragility, such as osteoporosis.
SCLEROSTIN (SOST) is a Wnt signaling inhibitor and a negative regulator of bone formation. Sclerostin is primarily produced by osteocytes, which are bone cells embedded within the bone matrix.
Sclerostin and DKK-1 can be measured in human serum samples with an ELISA assay.
The WNT signaling pathway is a central regulator of bone development and regeneration. Functional alterations of WNT ligands and inhibitors are associated with a variety of bone diseases that affect bone fragility and result in a high medical and socioeconomic burden. Hence, this cellular pathway has emerged as a novel target for bone-protective therapies, e.g. in osteoporosis. Here, we investigated glycosaminoglycan (GAG) recognition by Dickkopf-1 (DKK1), a potent endogenous WNT inhibitor, and the underlying functional implications in order to develop WNT signaling regulators. In a multidisciplinary approach we applied in silico structure-based de novo design strategies and molecular dynamics simulations combined with synthetic chemistry and surface plasmon resonance spectroscopy to Rationally Engineer oligomeric Glycosaminoglycan derivatives (REGAG) with improved neutralizing properties for DKK1. In vitro and in vivo assays show that the GAG modification to obtain REGAG translated into increased WNT pathway activity and improved bone regeneration in a mouse calvaria defect model with critical size bone lesions. Importantly, the developed REGAG outperformed polymeric high-sulfated hyaluronan (sHA3) in enhancing bone healing up to 50% due to their improved DKK1 binding properties. Thus, rationally engineered GAG variants may represent an innovative strategy to develop novel therapeutic approaches for regenerative medicine