1
|
Afsar B, Afsar RE, Caliskan Y, Lentine KL. The Relationship between Sclerostin and Kidney Transplantation Mineral Bone Disorders: A Molecule of Controversies. Calcif Tissue Int 2024; 115:339-361. [PMID: 39078512 PMCID: PMC11405501 DOI: 10.1007/s00223-024-01261-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 07/06/2024] [Indexed: 07/31/2024]
Abstract
Kidney transplantation is the most effective treatment option for most patients with end-stage kidney disease due to reduced mortality, decreased cardiovascular events and increased quality of life compared to patients treated with dialysis. However, kidney transplantation is not devoid of both acute and chronic complications including mineral bone disorders (MBD) which are already present in patients with chronic kidney disease (CKD) before kidney transplantation. The natural history of MBD after kidney transplantation is variable and new markers are needed to define MBD after kidney transplantation. One of these promising molecules is sclerostin. The main action of sclerostin is to inhibit bone formation and mineralization by blocking osteoblast differentiation and function. In kidney transplant recipients (KTRs), various studies have shown that sclerostin is associated with graft function, bone parameters, vascular calcification, and arterial stiffness although non-uniformly. Furthermore, data for inhibition of sclerostin with monoclonal antibody romosozumab for treatment of osteoporosis is available for general population but not in KTRs which osteoporosis is highly prevalent. In this narrative review, we have summarized the studies investigating the change of sclerostin before and after kidney transplantation, the relationship between sclerostin and laboratory parameters, bone metabolism and vascular calcification in the context of kidney transplantation. We also pointed out the uncertainties, explained the causes of divergent findings and suggest further potential study topics regarding sclerostin in kidney transplantation.
Collapse
Affiliation(s)
- Baris Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, Turkey.
- Department of Nephrology, Saint Loui University, Saint Louis University Hospital, Saint Louis, MO, USA.
| | - Rengin Elsurer Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, Turkey
- Department of Nephrology, Saint Loui University, Saint Louis University Hospital, Saint Louis, MO, USA
| | - Yasar Caliskan
- Department of Nephrology, Saint Loui University, Saint Louis University Hospital, Saint Louis, MO, USA
| | - Krista L Lentine
- Department of Nephrology, Saint Loui University, Saint Louis University Hospital, Saint Louis, MO, USA
| |
Collapse
|
2
|
Lin Y, Mao L, Chen S, Zhou C. Serum sclerostin in vascular calcification in CKD: a meta-analysis. Ren Fail 2023; 45:2186151. [PMID: 36880646 PMCID: PMC10013495 DOI: 10.1080/0886022x.2023.2186151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
Vascular calcification (VC) is recognized as a predictor of all-cause and CVD mortality in chronic kidney disease (CKD). VC in CKD is possibly associated with serum sclerostin. The study systematically investigated the role of serum sclerostin in VC in CKD. Following the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols, a systematic search was performed of the PubMed, Cochrane Library, and EMBASE databases from inception to 11 November 2022, to identify relevant eligible studies. The data were retrieved, analyzed, and summarized. The hazard ratios (HRs) and odds ratios (ORs) with their corresponding confidence intervals (CIs) were derived and pooled. Thirteen reports (3125 patients) met the inclusion criteria and were included. Sclerostin was associated with the presence of VC (pooled OR = 2.75, 95%CI = 1.81-4.19, p < 0.01) and all-cause mortality (pooled HR = 1.22, 95%CI = 1.19-1.25, p < 0.01) among patients with CKD, but with a decreased risk of cardiovascular events (HR = 0.98, 95%CI = 0.97-1.00, p = 0.02). This meta-analysis suggests that serum sclerostin is associated with VC and all-cause mortality among patients with CKD.
Collapse
Affiliation(s)
- Yan Lin
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, China
| | - Liman Mao
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, China
| | - Siqi Chen
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, China
| | - Canxin Zhou
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, China
| |
Collapse
|
3
|
Koh HB, Ryu JH, Kim SS, Kim MG, Park JB, Kim CD, Kang KP, Ro H, Han SY, Huh KH, Yang J. Association between sclerostin levels and vascular outcomes in kidney transplantation patients. J Nephrol 2023; 36:2091-2109. [PMID: 37751127 DOI: 10.1007/s40620-023-01732-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 07/03/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND The impact of circulating sclerostin levels on vascular calcification has shown conflicting results depending on the target population and vascular anatomy. This study investigated the associations of sclerostin levels with vascular outcomes in kidney transplant patients. METHODS In a prospective observational study of the Korean Cohort Study for Outcome in Patients with Kidney Transplantation, 591 patients with serum sclerostin level data prior to transplantation were analyzed. The main predictor was the pre-transplant sclerostin level. Vascular outcomes were the abdominal aortic calcification score and brachial-ankle pulse wave velocity measured at pre-transplant screening and three and five years after kidney transplantation. RESULTS In linear regression analysis, sclerostin level positively correlated with changes in abdominal aortic calcification score between baseline and five years after kidney transplantation (coefficient of 0.73 [95% CI, 0.11-1.35] and 0.74 [95% CI, 0.06-1.42] for second and third tertiles, respectively, vs the first tertile). In a longitudinal analysis over five years, using generalized estimating equations, the coefficient of the interaction (sclerostin × time) was significant with a positive value, indicating that higher sclerostin levels were associated with faster increase in post-transplant abdominal aortic calcification score. Linear regression analysis revealed a positive association between pre-transplant sclerostin levels and changes in brachial-ankle pulse wave velocity (coefficient of 126.7 [95% CI, 35.6-217.8], third vs first tertile). Moreover, a significant interaction was identified between sclerostin levels and brachial-ankle pulse wave velocity at five years. CONCLUSIONS Elevated pre-transplant sclerostin levels are associated with the progression of post-transplant aortic calcifications and arterial stiffness.
Collapse
Affiliation(s)
- Hee Byung Koh
- Department of Internal Medicine, Catholic Kwandong University International Saint Mary's Hospital, Incheon, Republic of Korea
| | - Jung Hwa Ryu
- Department of Internal Medicine, Ewha Womans University Seoul Hospital, Seoul, Republic of Korea
| | - Seung-Seob Kim
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Myung-Gyu Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jae Berm Park
- Department of Surgery, Seoul Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Chan Duk Kim
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Kyung Pyo Kang
- Department of Internal Medicine, Research Institute of Clinical Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Han Ro
- Department of Internal Medicine, Gil Hospital, Gachon University, Incheon, Republic of Korea
| | - Seung-Yeup Han
- Department of Internal Medicine, Dongsan Medical Center, Keimyung University, Daegu, Republic of Korea
| | - Kyu Ha Huh
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jaeseok Yang
- Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
4
|
Hobson S, Arefin S, Witasp A, Hernandez L, Kublickiene K, Shiels PG, Stenvinkel P. Accelerated Vascular Aging in Chronic Kidney Disease: The Potential for Novel Therapies. Circ Res 2023; 132:950-969. [PMID: 37053277 DOI: 10.1161/circresaha.122.321751] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
The pathophysiology of vascular disease is linked to accelerated biological aging and a combination of genetic, lifestyle, biological, and environmental risk factors. Within the scenario of uncontrolled artery wall aging processes, CKD (chronic kidney disease) stands out as a valid model for detailed structural, functional, and molecular studies of this process. The cardiorenal syndrome relates to the detrimental bidirectional interplay between the kidney and the cardiovascular system. In addition to established risk factors, this group of patients is subjected to a plethora of other emerging vascular risk factors, such as inflammation, oxidative stress, mitochondrial dysfunction, vitamin K deficiency, cellular senescence, somatic mutations, epigenetic modifications, and increased apoptosis. A better understanding of the molecular mechanisms through which the uremic milieu triggers and maintains early vascular aging processes, has provided important new clues on inflammatory pathways and emerging risk factors alike, and to the altered behavior of cells in the arterial wall. Advances in the understanding of the biology of uremic early vascular aging opens avenues to novel pharmacological and nutritional therapeutic interventions. Such strategies hold promise to improve future prevention and treatment of early vascular aging not only in CKD but also in the elderly general population.
Collapse
Affiliation(s)
- S Hobson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| | - S Arefin
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| | - A Witasp
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| | - L Hernandez
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| | - K Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| | - P G Shiels
- School of Molecular Biosciences, MVLS, University of Glasgow, United Kingdom (P.G.S.)
| | - P Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden (S.H., S.A., A.W., L.H., K.K., P.S.)
| |
Collapse
|
5
|
Hobson S, Arefin S, Rahman A, Hernandez L, Ebert T, de Loor H, Evenepoel P, Stenvinkel P, Kublickiene K. Indoxyl Sulphate Retention Is Associated with Microvascular Endothelial Dysfunction after Kidney Transplantation. Int J Mol Sci 2023; 24:ijms24043640. [PMID: 36835051 PMCID: PMC9960432 DOI: 10.3390/ijms24043640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Kidney transplantation (KTx) is the preferred form of renal replacement therapy in chronic kidney disease (CKD) patients, owing to increased quality of life and reduced mortality when compared to chronic dialysis. Risk of cardiovascular disease is reduced after KTx; however, it is still a leading cause of death in this patient population. Thus, we aimed to investigate whether functional properties of the vasculature differed two years post-KTx (postKTx) compared to baseline (time of KTx). Using the EndoPAT device in 27 CKD patients undergoing living-donor KTx, we found that vessel stiffness significantly improved while endothelial function worsened postKTx vs. baseline. Furthermore, baseline serum indoxyl sulphate (IS), but not p-cresyl sulphate, was independently negatively associated with reactive hyperemia index, a marker of endothelial function, and independently positively associated with P-selectin postKTx. Finally, to better understand the functional effects of IS in vessels, we incubated human resistance arteries with IS overnight and performed wire myography experiments ex vivo. IS-incubated arteries showed reduced bradykinin-mediated endothelium-dependent relaxation compared to controls via reduced nitric oxide (NO) contribution. Endothelium-independent relaxation in response to NO donor sodium nitroprusside was similar between IS and control groups. Together, our data suggest that IS promotes worsened endothelial dysfunction postKTx, which may contribute to the sustained CVD risk.
Collapse
Affiliation(s)
- Sam Hobson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska, Institutet, 141 52 Stockholm, Sweden
| | - Samsul Arefin
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska, Institutet, 141 52 Stockholm, Sweden
| | - Awahan Rahman
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska, Institutet, 141 52 Stockholm, Sweden
| | - Leah Hernandez
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska, Institutet, 141 52 Stockholm, Sweden
| | - Thomas Ebert
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska, Institutet, 141 52 Stockholm, Sweden
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, D-04103 Leipzig, Germany
| | - Henriette de Loor
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, Katholieke Universiteit Leuven, BE-3000 Leuven, Belgium
| | - Pieter Evenepoel
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, Katholieke Universiteit Leuven, BE-3000 Leuven, Belgium
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, BE-3000 Leuven, Belgium
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska, Institutet, 141 52 Stockholm, Sweden
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska, Institutet, 141 52 Stockholm, Sweden
- Correspondence:
| |
Collapse
|
6
|
Žuža I, Dodig D, Brumini I, Tokmadžić D, Orlić L, Zgrablić D, Vukelić I, Gršković A, Katalinić N, Jakšić A, Miletić D, Rački S, Markić D. A CT-based pelvic calcification score in kidney transplant patients is a possible predictor of graft and overall survival. Br J Radiol 2022; 95:20220394. [PMID: 36116132 PMCID: PMC9793470 DOI: 10.1259/bjr.20220394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/13/2022] [Accepted: 08/24/2022] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVES Computerized tomography (CT) is the most accurate method for evaluating pelvic calcifications, which are of utmost importance for planning kidney transplantation (KT). The aim of our study was to evaluate the incidence and distribution of iliac artery calcifications and correlate the novel pelvic calcification score (PCS) with cardiovascular risk factors and graft and overall survival in KT patients. METHODS We retrospectively included 118 KT patients operated at our institution with pretransplant pelvic CT. Calcification morphology, circumference and length of both common and external iliac arteries were independently scored by two uroradiologists. PCS was calculated as the total score sum of all three calcification features in all vessels. PCS correlation with graft and patient survival was performed. RESULTS Calcification in at least one vascular segment was found in 79% of patients. PCS was significantly higher in male patients (p = 0.006), patients over 55 years (p < 0.001), and patients on haemodialysis (p = 0.016). Patients with a PCS >3 had significantly shorter graft and overall survival rates (p = 0.041 and p = 0.039, respectively). CONCLUSIONS The extent of iliac artery calcification in KT recipients quantified by PCS on pretransplant CT correlates with graft and overall patient survival. A PCS over three was associated with worse clinical outcomes and could become a possible prognostic factor. ADVANCES IN KNOWLEDGE Our novel PCS is a robust method for quantifying iliac artery calcification burden. Since higher a PCS correlates with worse patient and graft survival, PCS has the potential to become a prognostic factor in kidney transplant patients.
Collapse
Affiliation(s)
- Iva Žuža
- Department of Radiology, University Hospital Rijeka, Rijeka, Croatia
| | - Doris Dodig
- Department of Radiology, University Hospital Rijeka, Rijeka, Croatia
| | - Ivan Brumini
- Department of Radiology, University Hospital Rijeka, Rijeka, Croatia
| | | | | | - David Zgrablić
- Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ivan Vukelić
- Department of Urology, University Hospital Rijeka, Rijeka, Croatia
| | | | | | - Ante Jakšić
- Department of Urology, University Hospital Rijeka, Rijeka, Croatia
| | | | | | | |
Collapse
|
7
|
Vascular Calcification Is Associated with Fetuin-A and Cortical Bone Porosity in Stone Formers. J Pers Med 2022; 12:jpm12071120. [PMID: 35887617 PMCID: PMC9319706 DOI: 10.3390/jpm12071120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Nephrolithiasis has been associated with bone loss and vascular calcification (VC), reflecting abnormal extraosseous calcium deposition. Fetuin-A (Fet-A) acts as a potent inhibitor of ectopic mineralization. The aim of the present study was to evaluate the prevalence of VC in stone formers (SF) and non-stone formers (NSF) and to investigate potential determinants of VC among SF, including circulating levels of Fet-A and bone microarchitecture parameters. Methods: Abdominal aortic calcification (AAC) was assessed using available computed tomography in SF and in age-, sex-, and BMI-matched NSF (potential living kidney donors). Serum Fet-A was measured in stored blood samples from SF. Bone microarchitecture parameters were obtained as a post hoc analysis of a cross-sectional cohort from young SF evaluated by high-resolution peripheral quantitative computed tomography (HR-pQCT). Results: A total of 62 SF (38.0 [28.0−45.3] years old) and 80 NSF (40.0 [37.0−45.8] years old) were included. There was no significant difference in AAC scores between SF and NSF. However, when dividing SF according to mean AAC score, below <5.8% (n = 33) or above ≥5.8% (n = 29), SF with higher AAC presented significantly higher BMI and tibial cortical porosity (Ct.Po) and significantly lower serum HDL, klotho, Fet-A, and eGFR. Urinary calcium did not differ between groups, but fractional excretion of phosphate was higher in the former. Upon multivariate regression, BMI, serum Fet-A, and tibial Ct.Po remained independently associated with AAC. Conclusions: This study suggests an association between reduced circulating Fet-A levels and increased bone Ct.Po with VC in SF.
Collapse
|
8
|
Abstract
Sclerostin is most recognized for its role in controlling bone formation but is also expressed in the heart, aorta, coronary, and peripheral arteries. This review summarizes research on sclerostin's role in cardiovascular disease. Rodent studies have found sclerostin to be expressed at sites of arterial calcification. In contrast, aortic sclerostin was reported to be downregulated in a mouse model of abdominal aortic aneurysm, and transgenic upregulation or administration of sclerostin was found to prevent abdominal aortic aneurysm and atherosclerosis formation. Sclerostin deficiency was reported to stimulate cardiac rupture in one rodent model. In humans, 7 of 11 studies reported a significant association between high serum sclerostin and high carotid intima media thickness. Ten of 15 studies reported a significant association between high serum sclerostin and severe arterial calcification. Twelve of 14 studies reported a significant association between high serum sclerostin and high arterial stiffness or atherosclerosis severity. Four of 9 studies reported a significant association between high serum sclerostin and high risk of cardiovascular events. A meta-analysis of randomized controlled trials suggested that administration of the sclerostin blocking antibody romosozumab did not significantly increase the risk of major adverse cardiovascular events (risk ratio, 1.14 [95% CI, 0.83-1.57]; P=0.54) or cardiovascular death (risk ratio, 0.92 [95% CI, 0.53-1.59]; P=0.71). Human genetic studies reported variants predisposing to low arterial sclerostin expression were associated with a high risk of cardiovascular events. Overall, past research suggests a cardiovascular protective role of sclerostin but findings have been inconsistent, possibly due to variations in study design, the unique populations and models studied, and the heterogeneous methods used.
Collapse
Affiliation(s)
- Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry (J.G., S.T.), James Cook University, Townsville, Queensland, Australia.,The Australian Institute of Tropical Health and Medicine J.G.' S.T.), James Cook University, Townsville, Queensland, Australia
| | - Shivshankar Thanigaimani
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry (J.G., S.T.), James Cook University, Townsville, Queensland, Australia.,The Australian Institute of Tropical Health and Medicine J.G.' S.T.), James Cook University, Townsville, Queensland, Australia.,The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Queensland, Australia (J.G.)
| |
Collapse
|
9
|
Erlandsson H, Qureshi AR, Ripsweden J, Haugen Löfman I, Söderberg M, Wennberg L, Lundgren T, Bruchfeld A, Brismar TB, Stenvinkel P. Scoring of medial arterial calcification predicts cardiovascular events and mortality after kidney transplantation. J Intern Med 2022; 291:813-823. [PMID: 35112417 PMCID: PMC9306575 DOI: 10.1111/joim.13459] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Progression of vascular calcification causes cardiovascular disease, which is the most common cause of death in chronic kidney failure and after kidney transplantation (KT). The prognostic impact of the extent of medial vascular calcification at KT is unknown. METHODS In this prospective cohort study, we investigated the impact of medial calcification compared to a mix of intimal and medial calcification represented by coronary artery calcification (CAC score) and aortic valve calcification in 342 patients starting on kidney failure replacement therapy. The primary outcomes were cardiovascular events (CVE) and death. The median follow-up time was 6.4 years (interquartile range 3.7-9.6 years). Exposure was CAC score and arteria epigastrica medial calcification scored as none, mild, moderate, or severe by a pathologist at time of KT (n = 200). We divided the patients according to kidney failure replacement therapy during follow-up, that is, living donor KT, deceased donor KT, or dialysis. RESULTS Moderate to severe medial calcification in the arteria epigastrica was associated with higher mortality (p = 0.001), and the hazard ratio for CVE was 3.1 (95% confidence interval [CI] 1.12-9.02, p < 0.05) compared to no or mild medial calcification. The hazard ratio for 10-year mortality in the dialysis group was 33.6 (95% CI, 10.0-113.0, p < 0.001) compared to living donor recipients, independent of Framingham risk score and prevalent CAC. CONCLUSION Scoring of medial calcification in the arteria epigastrica identified living donor recipients as having 3.1 times higher risk of CVE, independent of traditional risk factors. The medial calcification score could be a reliable method to identify patients with high and low risk of CVE and mortality following KT.
Collapse
Affiliation(s)
- Helen Erlandsson
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Abdul Rashid Qureshi
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Jonaz Ripsweden
- Department of Radiology, Karolinska University Hospital, Stockholm, Sweden.,Unit of radiology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Ida Haugen Löfman
- Section of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Söderberg
- Cardiovascular, Renal and Metabolism Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Lars Wennberg
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Torbjörn Lundgren
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Annette Bruchfeld
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Torkel B Brismar
- Department of Radiology, Karolinska University Hospital, Stockholm, Sweden.,Unit of radiology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden.,Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Kelly SP, Ramkumar DB, Peacock ZS, Newman ET, Venrick C, Lozano-Calderon SA, Raskin KA, Chebib I, Schwab JH. Sclerostin immunohistochemical staining in surgically treated giant cell tumor of bone. J Surg Oncol 2022; 126:571-576. [PMID: 35446992 DOI: 10.1002/jso.26903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/29/2022] [Accepted: 04/10/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Giant cell tumor of bone (GCTB) is a destructive lesion with a high potential for recurrence. RANK-ligand targeted therapy has provided promising, yet mixed results. Sclerostin (SOST) inhibition results in a net anabolic response and is currently used in the treatment of osteoporosis. The application to GCTB is unknown. OBJECTIVES We sought to determine if GCTB stained for SOST on immunohistochemistry and correlate its expression with predictor variables. METHODS All patients at a single institution undergoing surgery for GCTB between 1993 and 2008 with a minimum of 6 months follow-up were included. Primary outcomes included the presence of SOST staining, secondary outcomes included the correlation of patient and tumor-specific predictor variables. RESULTS SOST antibody staining of any cell type was present in 47 of 48 cases (97.9%). Positivity of the stromal cells was present in 39 of 48 cases (81.3%) and was associated with radiographic aggressiveness (p = 0.023), symptomatic presentation (p = 0.032), prior surgery (p = 0.005), and patient age (p = 0.034). Positivity of giant cells was present in 41 of 48 cases (85.4%) and was not significant with predictive factors. CONCLUSIONS Sclerostin staining in GCTB is a novel finding and warrants further research to define the role of sclerostin as a prognostic factor and therapeutic target.
Collapse
Affiliation(s)
- Sean P Kelly
- Department of Orthopaedic Surgery, Tripler Army Medical Center, Honolulu, Hawaii, USA
| | - Dipak B Ramkumar
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Orthopaedic Surgery, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Zachary S Peacock
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Erik T Newman
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Connor Venrick
- Department of Orthopaedic Surgery, Tripler Army Medical Center, Honolulu, Hawaii, USA
| | | | - Kevin A Raskin
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ivan Chebib
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joseph H Schwab
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Hasparyk UG, Vigil FMB, Bartolomei VS, Nunes VM, Simões e Silva AC. Chronic Kidney Disease-Mineral Bone Disease biomarkers in kidney transplant patients. Curr Med Chem 2022; 29:5230-5253. [DOI: 10.2174/0929867329666220318105856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 11/22/2022]
Abstract
Background:
Chronic Kidney Disease associated with Mineral Bone Disease (CKD-MBD) is frequent in kidney transplant patients. Post-transplantation bone disease is complex, especially in patients with pre-existing metabolic bone disorders that are further affected by immunosuppressive medications and changes in renal allograft function. Main biochemical abnormalities of mineral metabolism in kidney transplantation (KTx) include hypophosphatemia, hyperparathyroidism (HPTH), insufficiency or deficiency of vitamin D, and hypercalcemia.
Objective:
This review aimed to summarize the pathophysiology and main biomarkers of CKD-MBD in KTx.
Methods:
A comprehensive and non-systematic search in PubMed was independently made with an emphasis on biomarkers in mineral bone disease in KTx.
Results:
CKD-MBD can be associated with numerous factors including secondary HPTH, metabolic dysregulations before KTx, and glucocorticoids therapy in post-transplant subjects. Fibroblast growth factor 23 (FGF23) reaches normal levels after KTx with good allograft function, while calcium, vitamin D and phosphorus, ultimately, result in hypercalcemia, persistent vitamin D insufficiency, and hypophosphatemia respectively. As for PTH levels, there is an initial tendency of a significant decrease, followed by a raise due to secondary or tertiary HPTH. In regard to sclerostin levels, there is no consensus in the literature.
Conclusion:
KTx patients should be continuously evaluated for mineral homeostasis and bone status, both cases with successful kidney transplantation and those with reduced functionality. Additional research on CKD-MBD pathophysiology, diagnosis, and management is essential to guarantee long-term graft function, better prognosis, good quality of life, and reduced mortality for KTx patients.
Collapse
Affiliation(s)
- Ursula Gramiscelli Hasparyk
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Flávia Maria Borges Vigil
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Victória Soares Bartolomei
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Vitor Moreira Nunes
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ana Cristina Simões e Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
12
|
Van Berkel B, Van Ongeval C, Van Craenenbroeck AH, Pottel H, De Vusser K, Evenepoel P. Prevalence, progression and implications of breast artery calcification in patients with chronic kidney disease. Clin Kidney J 2022; 15:295-302. [PMID: 35145644 PMCID: PMC8825218 DOI: 10.1093/ckj/sfab178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Indexed: 12/19/2022] Open
Abstract
Breast arterial calcification (BAC) is increasingly recognized as a specific marker of medial calcification. The present retrospective observational cohort study aimed to define the prevalence, progression rate, risk factors and clinical implications of BAC in chronic kidney disease (CKD) patients across stages of disease. The presence and extent of BAC were determined on mammograms in 310 females (58.7 ± 10.8 years, Caucasian) with CKD across various stages of disease [CKD G2-5D n = 132; transplant (Tx) recipients n = 178]. In a subset of 88 patients, repeat mammography was performed, allowing us to calculate the annualized BAC rate. Overall, BAC was observed in 34.7% of the patients. BAC prevalence (P = 0.02) and BAC score (P = 0.05) increased along the progression of CKD. In the overall cohort, patients with BAC were characterized by older age, more cardiovascular disease, more inflammation, higher pulse pressure and borderline higher prevalence of diabetes and were more often treated with a vitamin K antagonist (VKA). The BAC progression rate was significantly lower in Tx patients as compared with CKD G5D. Progressors were characterized by more inflammation, worse kidney function, higher BAC score and higher serum phosphate level (Tx only) at baseline and were more often treated with a VKA. Major adverse cardiovascular event-free survival was significantly worse in Tx patients with BAC. In conclusion, BAC is common among CKD patients, progresses at a slower pace in Tx patients as compared with CKD 5D and associates with dismal cardiovascular outcomes. BAC score, kidney function, serum phosphate at baseline and VKA usage seem to be important determinants of progression.
Collapse
Affiliation(s)
- Brecht Van Berkel
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven and Laboratory of Nephrology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | - Chantal Van Ongeval
- Department of Imaging and Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Amaryllis H Van Craenenbroeck
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven and Laboratory of Nephrology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Kulak, Kortrijk, Belgium
| | - Katrien De Vusser
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven and Laboratory of Nephrology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | - Pieter Evenepoel
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven and Laboratory of Nephrology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Sclerostin Immunohistochemical Staining in Aggressive Maxillofacial Giant Cell Lesions: Initial Results and Potential Therapeutic Target. J Craniofac Surg 2021; 33:790-793. [PMID: 34753866 DOI: 10.1097/scs.0000000000008344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Maxillofacial (MF) giant cell lesions (GCLs) are benign, often locally aggressive lesions with potential for recurrence. Systemic treatments have included interferon alpha, calcitonin, bisphosphonates, and denosumab. Sclerostin (SOST) is typically thought to be a negative regulator of bone metabolism and anti-SOST agents have been used to treat osteoporosis; however, its role in central giant cell granuloma is unknown. The purpose of this study was to evaluate the expression of SOST in MF GCLs. MATERIALS AND METHODS This was a retrospective study of patients with MF GCLs treated at a single institution between 1993 and 2008 with a minimum follow-up of 6 months. Representative tissue was used to create a tissue microarray and SOST immunohistochemical (IHC) staining and grading was performed. The primary outcomes were IHC staining of the stromal cells and giant cells. The secondary outcomes included correlation of IHC staining and patient predictor variables including clinically benign and aggressive lesions. All analyses were completed using univariate statistical tests. RESULTS A total of 37 subjects were included (29 clinically aggressive and 8 clinically benign). Sclerostin staining was present in 30 of 37 subjects (81%). Of these, 22 (60%) had stromal cell staining and 28 (76%) had giant cell staining. The presence or absence of staining, of either cell type, was not associated with aggressiveness, presence of clinical symptoms, tumor size, previous interferon therapy, previous surgery, or the race or age of the patient. DISCUSSION Maxillofacial GCLs have an overall high level of SOST staining; however, the role of SOST in treatment and prognosis is unknown and warrants further study.
Collapse
|
14
|
Wu PY, Lee SY, Chang KV, Chao CT, Huang JW. Gender-Related Differences in Chronic Kidney Disease-Associated Vascular Calcification Risk and Potential Risk Mediators: A Scoping Review. Healthcare (Basel) 2021; 9:healthcare9080979. [PMID: 34442116 PMCID: PMC8394860 DOI: 10.3390/healthcare9080979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Vascular calcification (VC) involves the deposition of calcium apatite in vascular intima or media. Individuals of advanced age, having diabetes mellitus or chronic kidney disease (CKD) are particularly at risk. The pathogenesis of CKD-associated VC evolves considerably. The core driver is the phenotypic change involving vascular wall constituent cells toward manifestations similar to that undergone by osteoblasts. Gender-related differences are observed regarding the expressions of osteogenesis-regulating effectors, and presumably the prevalence/risk of CKD-associated VC exhibits gender-related differences as well. Despite the wealth of data focusing on gender-related differences in the risk of atherosclerosis, few report whether gender modifies the risk of VC, especially CKD-associated cases. We systematically identified studies of CKD-associated VC or its regulators/modifiers reporting data about gender distributions, and extracted results from 167 articles. A significantly higher risk of CKD-associated VC was observed in males among the majority of original investigations. However, substantial heterogeneity exists, since multiple large-scale studies yielded neutral findings. Differences in gender-related VC risk may result from variations in VC assessment methods, the anatomical segments of interest, study sample size, and even the ethnic origins of participants. From a biological perspective, plausible mediators of gender-related VC differences include body composition discrepancies, alterations involving lipid profiles, inflammatory severity, diversities in matrix Gla protein (MGP), soluble Klotho, vitamin D, sclerostin, parathyroid hormone (PTH), fibroblast growth factor-23 (FGF-23), and osteoprotegerin levels. Based on our findings, it may be inappropriate to monotonously assume that male patients with CKD are at risk of VC compared to females, and we should consider more background in context before result interpretation.
Collapse
Affiliation(s)
- Patrick Yihong Wu
- School of Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan;
| | - Szu-Ying Lee
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin County 640, Taiwan; (S.-Y.L.); (J.-W.H.)
| | - Ke-Vin Chang
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan;
| | - Chia-Ter Chao
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital BeiHu Branch, Taipei 10845, Taiwan
- Correspondence: ; Tel.: +886-2-23717101 (ext. 6531); Fax: +886-2-23717101
| | - Jenq-Wen Huang
- Nephrology Division, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin County 640, Taiwan; (S.-Y.L.); (J.-W.H.)
- Nephrology Division, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| |
Collapse
|
15
|
Abstract
The incidence of osteoporosis and cardiovascular disease increases with age, and there are potentially shared mechanistic associations between the two conditions. It is therefore highly relevant to understand the cardiovascular implications of osteoporosis medications. These are presented in this narrative review. Calcium supplementation could theoretically cause atheroma formation via calcium deposition, and in one study was found to be associated with myocardial infarction, but this has not been replicated. Vitamin D supplementation has been extensively investigated for cardiac benefit, but no consistent effect has been found. Despite findings in the early 21st century that menopausal hormone therapy was associated with coronary artery disease and venous thromboembolism (VTE), this therapy is now thought to be potentially safe (from a cardiac perspective) if started within the first 10 years of the menopause. Selective estrogen receptor modulators (SERMs) are associated with increased risk of VTE and may be related to fatal strokes (a subset of total strokes). Bisphosphonates could theoretically provide protection against atheroma. However, data from randomised trials and observational studies have neither robustly supported this nor consistently demonstrated the potential association with atrial fibrillation. Denosumab does not appear to be associated with cardiovascular disease and, although parathyroid hormone analogues are associated with palpitations and dizziness, no association with a defined cardiovascular pathology has been demonstrated. Finally, romosozumab has been shown to have a possible cardiovascular signal, and therefore post-market surveillance of this therapy will be vital.
Collapse
|
16
|
Fehérvári L, Frigy A, Kocsis L, Szabó IA, Szabo TM, Urkon M, Jakó Z, Nagy EE. Serum Osteoprotegerin and Carotid Intima-Media Thickness Are Related to High Arterial Stiffness in Heart Failure with Reduced Ejection Fraction. Diagnostics (Basel) 2021; 11:diagnostics11050764. [PMID: 33923139 PMCID: PMC8145213 DOI: 10.3390/diagnostics11050764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
Arterial stiffness (AS) is a complex vascular phenomenon with consequences for central hemodynamics and left-ventricular performance. Circulating biomarkers have been associated with AS; however, their value in heart failure is poorly characterized. Our aim was to evaluate the clinical and biomarker correlates of AS in the setting of heart failure with reduced ejection fraction (HFrEF). In 78 hospitalized, hemodynamically stable patients (20 women, 58 men, mean age 65.8 ± 1.41 years) with HFrEF, AS was measured using aortic pulse wave velocity (PWV). Serum OPG, RANKL, sclerostin, and DKK-1 were determined, and the relationships between the clinical variables, vascular-calcification-related biomarkers, and PWV were evaluated by correlation analysis and linear and logistic regression models. OPG and the OPG/RANKL ratio were significantly higher in the group of patients (n = 37, 47.4%) with increased PWV (>10 m/s). PWV was positively correlated with age, left-ventricular ejection fraction, and carotid intima-media thickness (cIMT), and negatively correlated with the glomerular filtration rate. OPG and cIMT were significantly associated with PWV in the logistic regression models when adjusted for hypertension, EF, and the presence of atherosclerotic manifestations. Elevated serum OPG, together with cIMT, were significantly related to increased AS in the setting of HFrEF.
Collapse
Affiliation(s)
- Lajos Fehérvári
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Attila Frigy
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Lóránd Kocsis
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
| | - István Adorján Szabó
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Timea Magdolna Szabo
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Melinda Urkon
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Zita Jakó
- Laboratory of Medical Analysis, Emergency Clinical County Hospital Targu Mures, 540136 Targu Mures, Romania;
| | - Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Laboratory of Medical Analysis, Clinical County Hospital Mures, 540394 Targu Mures, Romania
- Correspondence: ; Tel.: +40-733-956-395
| |
Collapse
|
17
|
Ahn SH, Ko MM, Song JH, Jung JH. Changes in plasma sclerostin level associated with use of a medium cut-off dialyzer in end-stage renal disease. Kidney Res Clin Pract 2021; 40:120-134. [PMID: 33745263 PMCID: PMC8041631 DOI: 10.23876/j.krcp.20.173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/01/2020] [Indexed: 12/04/2022] Open
Abstract
Background Larger middle molecules are important substances associated with cardiovascular complications in end- stage renal disease. Unfortunately, larger middle molecules are not reliably removed by a high -flux dialyzer. A medium cut-off (MCO) membrane could effectively remove larger middle molecules. This study aimed to identify the long-term effect of the MCO membrane for changes of larger middle molecules. Methods Thirty-four patients were prospectively analyzed for 12 months. The enrolled patients were divided into control and MCO groups. We measured the plasma levels of growth differentiation factor 15, sclerostin, and fibroblast growth factor 23 in larger middle molecules and those of biomarkers including small solutes. Single-pool Kt/V (spKt/V) and reduction ratios also were evaluated. Results Plasma sclerostin did not increase significantly in patients using the MCO dialyzer (135.294 [–637.726 to 908.314], p = 0.715). And there was a significant difference in change of plasma sclerostin level between the two groups (–1,646.916 [–3,015.150 to –278.682], p = 0.033). Furthermore, a negative association between calcium and sclerostin was not observed in the MCO group (r = –0.142, p = 0.587). Solute clearance of larger middle molecules in the MCO group was significantly higher. Moreover, spKt/V values for patients in the MCO group were significantly increased without albumin loss. Values are presented as mean (95% confidence interval [CI]) or adjusted mean (95% CI). Conclusion The MCO dialyzer can increase dialytic adequacy and suppress the increase in plasma sclerostin level without significant albumin loss in patients with end-stage renal disease.
Collapse
Affiliation(s)
- Seon-Ho Ahn
- Division of Nephrology, Department of Internal Medicine, Wonkwang University Hospital, Wonkwang University School of Medicine Hospital, Iksan, Republic of Korea
| | - Mi Mi Ko
- KM Fundamental Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Ju Hung Song
- Division of Nephrology, Department of Internal Medicine, Wonkwang University Hospital, Wonkwang University School of Medicine Hospital, Iksan, Republic of Korea
| | - Jong Hwan Jung
- Division of Nephrology, Department of Internal Medicine, Wonkwang University Hospital, Wonkwang University School of Medicine Hospital, Iksan, Republic of Korea
| |
Collapse
|
18
|
Alappan HR, Vasanth P, Manzoor S, O'Neill WC. Vascular Calcification Slows But Does Not Regress After Kidney Transplantation. Kidney Int Rep 2020; 5:2212-2217. [PMID: 33305114 PMCID: PMC7710884 DOI: 10.1016/j.ekir.2020.09.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Medial arterial calcification is a common and progressive lesion in end-stage renal disease that is associated with poor cardiovascular outcomes. Whether this lesion can be arrested or reversed is unknown, and was examined retrospectively by measuring progression of breast arterial calcification before and after kidney transplantation. Methods Arterial calcification was measured on serial mammograms from patients with previous kidney transplantation and compared to measurements performed before transplantation or in patients on the active waitlist. Serum creatinine >2.0 mg/dl after transplantation or warfarin use were exclusions. Results Median (interquartile range) progression of arterial calcification was 12.9 mm/breast per year (5.9 to 32.6) in 34 patients before or awaiting transplantation compared to just 1.2 mm/breast per year (-0.54 to 5.1) in 34 patients after transplantation (P < 0.001). Slowing of progression was also seen in longitudinal analyses of patients with mammograms performed both before and after transplantation. Duration of end-stage renal disease before transplantation but not age, diabetes, baseline calcification, or serum chemistries correlated with progression after transplantation. Significant regression was not observed in any patient. Conclusion In this first quantitative study of the effect of kidney transplantation, medial arterial calcification appeared to slow to rates seen in patients with normal renal function, indicating that the effect of renal failure may be completely abrogated. Overall, however, there was no significant regression, suggesting that calcification is irreversible and emphasizing the importance of prevention. Duration of pretransplant end-stage renal disease but not baseline calcification was a determinant of progression, consistent with cumulative, permanent changes to arteries that promote calcification.
Collapse
Affiliation(s)
- Harish R Alappan
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Payaswini Vasanth
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shumila Manzoor
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - W Charles O'Neill
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
19
|
Zeng S, Slowinski T, Pommer W, Hasan AA, Gaballa MMS, Lu Y, Krämer BK, Hocher B. Sclerostin is an independent risk factor for all-cause mortality in kidney transplant recipients. Clin Exp Nephrol 2020; 24:1177-1183. [PMID: 32816133 PMCID: PMC7599189 DOI: 10.1007/s10157-020-01956-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/07/2020] [Indexed: 12/31/2022]
Abstract
Background Sclerostin is a hormone contributing to the bone-vascular wall cross talk and has been implicated in cardiovascular events and mortality in patients with chronic kidney disease (CKD). We analyzed the relationship between sclerostin and mortality in renal transplant recipients. Methods 600 stable renal transplant recipients (367men, 233 women) were followed for all-cause mortality for 3 years. Blood and urine samples for analysis and clinical data were collected at study entry. We performed Kaplan–Meier survival analysis and Cox regression models considering confounding factors such as age, eGFR, cold ischemia time, HbA1c, phosphate, calcium, and albumin. Optimal cut-off values for the Cox regression model were calculated based on ROC analysis. Results Sixty-five patients died during the observation period. Nonsurvivors (n = 65; sclerostin 57.31 ± 30.28 pmol/L) had higher plasma sclerostin levels than survivors (n = 535; sclerostin 47.52 ± 24.87 pmol/L) (p = 0.0036). Kaplan–Meier curve showed that baseline plasma sclerostin concentrations were associated with all-cause mortality in stable kidney transplant recipients (p = 0.0085, log-rank test). After multiple Cox regression analysis, plasma levels of sclerostin remained an independent predictor of all-cause mortality (hazard ratio, 1.011; 95% CI 1.002–1.020; p = 0.0137). Conclusions Baseline plasma sclerostin is an independent risk factor for all-cause mortality in patients after kidney transplantation. Electronic supplementary material The online version of this article (10.1007/s10157-020-01956-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shufei Zeng
- Department of Nephrology, Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany.,Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Torsten Slowinski
- Department of Nephrology, Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Wolfgang Pommer
- KfH Kuratorium für Dialyse und Nierentransplantation e.V., Bildungszentrum, Neu-Isenburg, Germany
| | - Ahmed A Hasan
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Mohamed M S Gaballa
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Institute of Nutritional Science, University of Potsdam, Potsdam, Germany.,Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Egypt
| | - Yongping Lu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany. .,Institute of Medical Diagnostics, IMD Berlin, Berlin, Germany. .,Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China. .,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China.
| |
Collapse
|
20
|
Candellier A, Hénaut L, Morelle J, Choukroun G, Jadoul M, Brazier M, Goffin É. Aortic stenosis in patients with kidney failure: Is there an advantage for a PD-first policy? Perit Dial Int 2020; 41:158-167. [DOI: 10.1177/0896860820941371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Aortic stenosis (AS) is the most common valvular disease. It is twice as prevalent in patients with kidney failure as compared to the general population. In addition, AS progresses at a faster rate and is associated with a higher risk of death and poorer quality of life in patients on dialysis. Chronic kidney disease–mineral and bone disorder (CKD-MBD), inflammation, and hemodynamic disturbances contribute to the pathophysiology and progression of AS. Whether the type of dialysis modality, that is, hemodialysis (HD) versus peritoneal dialysis (PD), has a differential impact on the development and progression of AS in patients with kidney failure remains debated. Recent data indicate that the prevalence of valvular calcifications might be lower and the development of AS delayed in PD patients, as compared to those treated with HD. This could be accounted for by several mechanisms including reduced valvular shear stress, better preservation of residual kidney function (with better removal of protein-bound uremic toxins and CKD-MBD profile), and lower levels of systemic inflammation. Given the high morbidity and mortality rates related to interventional procedures in the population with kidney failure, surgical and transcatheter aortic valve replacement should be considered in selected patients with severe AS. Strategies slowing down the progression of aortic valve remodeling should remain the cornerstone in the management of individuals with kidney failure and mild to moderate AS. This review explores the potential benefits of PD in patients with kidney failure and AS and provides some clues to help clinicians in the decision-making process when options for kidney replacement therapy are considered in patients with AS.
Collapse
Affiliation(s)
- Alexandre Candellier
- Division of Nephrology, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
- UR UPJV 7517, MP3CV, CURS, Amiens, France
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | | | - Johann Morelle
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Gabriel Choukroun
- Division of Nephrology, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
- UR UPJV 7517, MP3CV, CURS, Amiens, France
| | - Michel Jadoul
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | | | - Éric Goffin
- Division of Nephrology, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| |
Collapse
|
21
|
Pereira L, Frazão JM. The bone-vessel axis in chronic kidney disease: An update on biochemical players and its future role in laboratory medicine. Clin Chim Acta 2020; 508:221-227. [PMID: 32422129 DOI: 10.1016/j.cca.2020.05.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/31/2020] [Accepted: 05/11/2020] [Indexed: 12/16/2022]
Abstract
Vascular wall calcification (VC) is highly prevalent in patients with chronic kidney disease (CKD). In CKD, VC is more frequent and severe than in the general population and it is associated with increased cardiovascular mortality and morbidity. In the last years, laboratory and clinical evidence have drawn the attention to the relationship between bone disease and VC in CKD patients, leading to the concept of a bone-vessel or bone-vascular axis. It means that disorders of bone volume and bone turnover may influence the risk of VC and ultimately the high risk of cardiovascular mortality. In fact, a higher burden of VC has been associated to low bone volume and low bone turnover in hemodialysis (HD) patients with renal osteodystrophy characterized by histomorphometric evaluation of bone biopsies. The molecular mechanisms underlying the regulation of bone cells and vascular cells in CKD are poorly understood. In this review, we discuss relevant evidence linking bone disorders and VC in CKD and also rising molecular players involved in this bone-vascular axis. Indeed, accumulating data is available for two proposed systems: receptor activator for nuclear factor kB (RANK)/ RANK ligand (RANKL)/osteoprotegerin (OPG) system and inhibitors of Wnt signaling - mainly sclerostin. Although they are promising biochemical markers linking bone formation and bone reabsorption with VC, there is a long way to go as long evidence from laboratory studies is often divergent to the clinical data as will be discussed. Future prospective studies are needed in order to evaluate the role of these biochemical players as useful clinical markers for VC, bone volume and perhaps bone turnover.
Collapse
Affiliation(s)
- Luciano Pereira
- Institute of Investigation and Innovation in Health, University of Porto, Portugal; INEB - National Institute of Biomedical Engineering, University of Porto, Portugal; Department of Nephrology, São João Hospital Center, Porto, Portugal
| | - João M Frazão
- Institute of Investigation and Innovation in Health, University of Porto, Portugal; INEB - National Institute of Biomedical Engineering, University of Porto, Portugal; Department of Nephrology, São João Hospital Center, Porto, Portugal.
| |
Collapse
|
22
|
De Maré A, D’Haese PC, Verhulst A. The Role of Sclerostin in Bone and Ectopic Calcification. Int J Mol Sci 2020; 21:ijms21093199. [PMID: 32366042 PMCID: PMC7246472 DOI: 10.3390/ijms21093199] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023] Open
Abstract
Sclerostin, a 22-kDa glycoprotein that is mainly secreted by the osteocytes, is a soluble inhibitor of canonical Wnt signaling. Therefore, when present at increased concentrations, it leads to an increased bone resorption and decreased bone formation. Serum sclerostin levels are known to be increased in the elderly and in patients with chronic kidney disease. In these patient populations, there is a high incidence of ectopic cardiovascular calcification. These calcifications are strongly associated with cardiovascular morbidity and mortality. Although data are still controversial, it is likely that there is a link between ectopic calcification and serum sclerostin levels. The main question, however, remains whether sclerostin exerts either a protective or deleterious role in the ectopic calcification process.
Collapse
|
23
|
Abstract
Over the last decades, the association between vascular calcification (VC) and all-cause/cardiovascular mortality, especially in patients with high atherogenic status, such as those with diabetes and/or chronic kidney disease, has been repeatedly highlighted. For over a century, VC has been noted as a passive, degenerative, aging process without any treatment options. However, during the past decades, studies confirmed that mineralization of the arteries is an active, complex process, similar to bone genesis and formation. The main purpose of this review is to provide an update of the existing biomarkers of VC in serum and develop the various pathogenetic mechanisms underlying the calcification process, including the pivotal roles of matrix Gla protein, osteoprotegerin, bone morphogenetic proteins, fetuin-a, fibroblast growth-factor-23, osteocalcin, osteopontin, osteonectin, sclerostin, pyrophosphate, Smads, fibrillin-1 and carbonic anhydrase II.
Collapse
|
24
|
Bouquegneau A, Evenepoel P, Paquot F, Malaise O, Cavalier E, Delanaye P. Sclerostin within the chronic kidney disease spectrum. Clin Chim Acta 2019; 502:84-90. [PMID: 31866333 DOI: 10.1016/j.cca.2019.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 01/08/2023]
Abstract
Sclerostin is sometimes presented as a promising biomarker in assessing bone health both in the general population and chronic kidney disease patients. However, it is still unclear whether it has any true added value compared to existing bone biomarkers in predicting bone turnover and/or bone density in chronic kidney disease patients. A wealth of papers has been published to evaluate the association between sclerostin and vascular calcifications development or even as prognostic biomarker for mortality, but often with conflicting results. Standardization and harmonization of analytical techniques is a prerequisite to advance clinical knowledge in sclerostin.
Collapse
Affiliation(s)
- Antoine Bouquegneau
- Department of Nephrology-Dialysis-Transplantation, University Hospital of Liege (ULg CHU), Liege, Belgium.
| | - Peter Evenepoel
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - François Paquot
- Department of Nephrology-Dialysis-Transplantation, University Hospital of Liege (ULg CHU), Liege, Belgium
| | - Olivier Malaise
- Department of Rheumatology, University Hospital of Liege (ULg CHU), Liege, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, University Hospital of Liege (ULg CHU), Liege, Belgium
| | - Pierre Delanaye
- Department of Nephrology-Dialysis-Transplantation, University Hospital of Liege (ULg CHU), Liege, Belgium
| |
Collapse
|
25
|
Abstract
The causes of the increased cardiovascular risk associated with kidney diseases partly reside in the chronic kidney disease-mineral bone disorder (CKD-MBD) syndrome. Three cardiovascular risk factors [hyperphosphatemia, vascular calcification, and elevated fibroblast growth factor 23 (FGF23)] levels have been discovered within the CKD-MBD over the last decades. In addition, sclerostin is recently presented as a new bone and vascular disease biomarker. This 22-kDa glycoprotein, secreted mainly by osteocytes, is a soluble inhibitor of the canonical Wnt pathway that has a pivotal role in bone biology and turnover. CKD patients are reported with higher levels of sclerostin, and levels decrease during dialysis. Sclerostin is associated with vascular calcification and CV risk in CKD, although data are still controversial. The question whether serum sclerostin has protective or deleterious role in CKD-MBD pathophysiology, and therefore in cardiovascular risk and overall mortality, is still open and needs to be answered. The standardization of assays and the establishment of a clear cut-off values when sclerostin starts to switch from physiological to pathophysiological role have to be another important step. Further research is needed also to define its relationship with other CKD-MBD biomarkers for future diagnostic and therapeutic strategies.
Collapse
|
26
|
Hassan MG, Morise F, Osman NA, Salam LA, Yehia H, Hamdi M, El Husseiny NM, NasrAllah MM. Micro RNA-192 Is Negatively Associated With Cardiovascular Events Among Wait-Listed Potential Kidney Transplant Recipients on Hemodialysis Over a 5-year Follow-up Period. Transplant Proc 2019; 51:2237-2240. [PMID: 31399202 DOI: 10.1016/j.transproceed.2019.02.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/29/2019] [Accepted: 02/17/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Patients with chronic renal disease are susceptible to accelerated vascular calcification and cardiovascular morbidity and mortality. Micro RNAs (miRNAs) have been linked to the pathogenesis of cardiovascular diseases in the general population. AIM This study was carried out to evaluate the link between miRNA 192 and vascular calcification, pre-existing as well as newly occurring major adverse cardiovascular events, and mortality among hemodialysis patients who are also considered to be potential kidney transplant recipients. METHODS We screened 64 potential transplant recipients on hemodialysis at our university hospital. Pre-existing overt cardiovascular disease was recorded; new adverse cardiovascular events and all causes of death over an observational period of 5 years were prospectively followed. Vascular calcification was measured in the aorta using computerized tomography scans, and micro RNA 192 was measured. RESULTS The final study population included 55 patients followed for 63 months. Micro RNA 192 was significantly lower in patients who had preexisting cardiovascular disease (P = .015) as well and in all patients who had experienced any event by the end of the observational period (P = .012). A multiregression analysis model including micro RNA, age, dialysis vintage, intradialytic hypotension, vascular calcification, diabetes, systolic blood pressure, and smoking found the only independently correlating factor to cardiovascular events in this model to be micro RNA (β = -0.286, P = .05). CONCLUSIONS MiRNA 192 levels are significantly lower among patients experiencing cardiovascular events while on hemodialysis awaiting kidney transplantation.
Collapse
Affiliation(s)
- Mona G Hassan
- Department of Nephrology & Department of Internal Medicine, Kasr Alainy School of Medicine, Cairo University, Cairo, Egypt
| | - Fadia Morise
- Department of Nephrology & Department of Internal Medicine, Kasr Alainy School of Medicine, Cairo University, Cairo, Egypt
| | - Noha A Osman
- Department of Nephrology, Kasr Alainy School of Medicine, Cairo University, Cairo, Egypt
| | - Lobna Abdel Salam
- Genome Unit, Kasr Alainy School of Medicine, Cairo University, Cairo, Egypt
| | - Hesham Yehia
- Department of Cardiology, Kasr Alainy School of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed Hamdi
- Department of Critical Care, Kasr Alainy School of Medicine, Cairo University, Cairo, Egypt
| | - Noha M El Husseiny
- Department of Internal Medicine, Kasr Alainy School of Medicine, Cairo University, Cairo, Egypt
| | - M M NasrAllah
- Department of Nephrology, Kasr Alainy School of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
27
|
Kalousová M, Dusilová-Sulková S, Kuběna AA, Zakiyanov O, Tesař V, Zima T. Sclerostin levels predict cardiovascular mortality in long-term hemodialysis patients: A prospective observational cohort study. Physiol Res 2019; 68:547-558. [PMID: 31177791 DOI: 10.33549/physiolres.934034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Sclerostin is a protein which is involved in bone metabolism and probably also in vessel wall function. This prospective observational cohort study evaluated the prognostic significance of sclerostin in hemodialysis (HD) patients. In total, 106 HD patients and 25 healthy controls participated in the study. HD patients were prospectively followed up for five years. Sclerostin was measured in serum using standard ELISA kits by Biomedica. Sclerostin concentrations in serum were higher in HD patients compared to the controls (89.2±40.3 pmol/l vs. 32.8±13.0 pmol/l, p<0.001). Sclerostin levels were significant for cardiovascular mortality but not for overall mortality and mortality due to infection. A higher cardiovascular risk was connected to sclerostin concentrations above the median (>84 pmol/l), HR (95 % CI): 2.577 (1.0002-10.207), p=0.04. When sclerostin was evaluated together with residual diuresis in Kaplan-Meier analysis the worst prognosis due to cardiovascular events was observed in the group with high sclerostin and zero residual diuresis compared to all other patients (p=0.007). In summary, serum sclerostin levels in HD patients were increased when compared to healthy subjects. High sclerostin levels were demonstrated as a risk factor for cardiovascular mortality. Further studies are required to clarify the pathophysiological mechanisms of sclerostin action in patients with renal failure before therapeutic measures can be established.
Collapse
Affiliation(s)
- M Kalousová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
28
|
Asadipooya K, Weinstock A. Cardiovascular Outcomes of Romosozumab and Protective Role of Alendronate. Arterioscler Thromb Vasc Biol 2019; 39:1343-1350. [PMID: 31242037 DOI: 10.1161/atvbaha.119.312371] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Osteoporosis and cardiovascular diseases are major public health issues. Bone and cardiovascular remodeling share multiple biological markers and pathways. Medical intervention, such as using romosozumab, an antisclerostin antibody, improves the clinical outcome of osteoporosis. However, blocking sclerostin leads to Wnt (wingless/integrated) activation and participation in the cardiovascular remodeling process, which could potentially lead to adverse events. Based on the opposing roles of bisphosphonates and the Wnt pathway on endothelial dysfunction, lipid accumulation and calcification of the vessel walls, the combination of romosozumab and bisphosphonates could be a new therapeutic approach to reducing the risks of adverse cardiovascular events in romosozumab receivers. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Kamyar Asadipooya
- From the Division of Endocrinology and Molecular Medicine, Department of Medicine, University of Kentucky, Lexington (K.A.)
| | - Ada Weinstock
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York (A.W.)
| |
Collapse
|
29
|
Li M, Zhou H, Yang M, Xing C. 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. Int Urol Nephrol 2018; 51:311-323. [PMID: 30515734 DOI: 10.1007/s11255-018-2033-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 11/19/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Vascular calcification (VC) is known to be prevalent in patients with end-stage renal disease (ESRD). Sclerostin has been identified to be involved in the cross-talk between the kidney, vasculature, and bone. The aims of the present study were to evaluate vessel sclerostin expression and its correlation with VC, as well as serum sclerostin levels. METHODS A total of 51 adult ESRD patients undergoing living donor renal transplant (RT) were enrolled in this study. Serum sclerostin levels were measured by enzyme-linked immunosorbent assays. The thoracic aorta calcification (TAC) was measured by computed tomography (CT). The aortic calcification area index (ACAI) was used to evaluate the severity of TAC. During the RT surgery, the internal iliac arteries were collected and paraffin-embedded in 40 patients, followed by immunohistochemical staining for sclerostin expression and von Kossa-staining for vascular medial calcification degree. RESULTS The prevalence rate of TAC detected by CT was 58.82%. The positive rates of the internal iliac arterial calcification and vessel sclerostin expression were both 45%. Vessel sclerostin was strongly co-localized with medial calcification. Multivariate analyses revealed that only serum sclerostin was significantly associated with the presence of TAC, the severity of TAC and the positive expression of vessel sclerostin. Kappa test showed that the consistency of the two different calcification assessment methods, as well as the consistency of vessel sclerostin expression and von Kossa-staining were high. Furthermore, the cutoff points of serum sclerostin for vessel sclerostin expression, the presence of VC evaluated by CT and that evaluated by pathology were 1599.92 pg/mL, 2475.52 pg/mL, and 2116.23 pg/mL, respectively. CONCLUSIONS The two methods, namely CT and pathology, to evaluate VC were highly consistent. Serum sclerostin was an independent determinant of positive expression of vessel sclerostin and VC in ESRD patients eligible for RT.
Collapse
Affiliation(s)
- Min Li
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.,Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Hua Zhou
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Min Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Changying Xing
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
30
|
Appelman-Dijkstra NM, Papapoulos SE. Clinical advantages and disadvantages of anabolic bone therapies targeting the WNT pathway. Nat Rev Endocrinol 2018; 14:605-623. [PMID: 30181608 DOI: 10.1038/s41574-018-0087-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The WNT signalling pathway is a key regulator of bone metabolism, particularly bone formation, which has helped to define the role of osteocytes - the most abundant bone cells - as orchestrators of bone remodelling. Several molecules involved in the control of the WNT signalling pathway have been identified as potential targets for the development of bone-building therapeutics for patients with osteoporosis. Several of these molecules have been investigated in animal models, but only inhibitors of sclerostin (which is produced by osteocytes) have been investigated in phase III clinical studies. Here, we review the rationale for these developments and the specificity and potential off-target actions of WNT-based therapeutics. We also describe the available preclinical and clinical studies and discuss the benefits and risks of using sclerostin inhibitors for the management of patients with osteoporosis.
Collapse
|
31
|
Lu CL, Shyu JF, Wu CC, Hung CF, Liao MT, Liu WC, Zheng CM, Hou YC, Lin YF, Lu KC. Association of Anabolic Effect of Calcitriol with Osteoclast-Derived Wnt 10b Secretion. Nutrients 2018; 10:nu10091164. [PMID: 30149605 PMCID: PMC6164019 DOI: 10.3390/nu10091164] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/21/2018] [Accepted: 08/23/2018] [Indexed: 01/02/2023] Open
Abstract
Canonical Wnt (Wingless/Integrated) signaling is crucial in bone development and the Wnt ligand can promote osteoblast differentiation from mesenchymal progenitor cells. Calcitriol, an active vitamin D3, is used clinically for treatment of secondary hyperparathyroidism (SHPT) in chronic kidney disease (CKD) patients. The bone effects of calcitriol in SHPT remains uncertain. We hypothesized that calcitriol improves bone mass by suppressing osteoclast activity, and simultaneously promoting Wnt ligand secretion. We designed a cross-sectional study in maintenance hemodialysis patients to explore the effects of calcitriol on different bone turnover markers and specifically emphasized the Wnt 10b levels. Then, we explored the source of Wnt 10b secretion by using osteoclasts and osteoblasts treated with calcitriol in cell culture studies. Finally, we explored the effects of calcitriol on bone microarchitectures in CKD mice, using the 5/6 nephrectomy CKD animal model with analysis using micro-computed tomography. Calcitriol promoted the growth of both trabecular and cortical bones in the CKD mice. Wnt 10b and Procollagen 1 N-terminal Propeptide (P1NP) significantly increased, but Tartrate-resistant acid phosphatase 5b (Trap 5b) significantly decreased in the calcitriol-treated maintenance hemodialysis patients. Calcitriol enhanced Wnt 10b secretion from osteoclasts in a dose-dependent manner. Treatment of SHPT with calcitriol improved the bone anabolism by inhibiting osteoclasts and promoting osteoblasts that might be achieved by increasing the Wnt 10b level.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Anabolic Agents/therapeutic use
- Animals
- Biomarkers/metabolism
- Bone Remodeling/drug effects
- Calcitriol/therapeutic use
- Cells, Cultured
- Cross-Sectional Studies
- Disease Models, Animal
- Female
- Humans
- Hyperparathyroidism, Secondary/drug therapy
- Hyperparathyroidism, Secondary/etiology
- Hyperparathyroidism, Secondary/metabolism
- Hyperparathyroidism, Secondary/physiopathology
- Male
- Mice, Inbred C57BL
- Middle Aged
- Osteoblasts/drug effects
- Osteoblasts/metabolism
- Osteoclasts/drug effects
- Osteoclasts/metabolism
- Proto-Oncogene Proteins/metabolism
- Rats, Sprague-Dawley
- Renal Dialysis
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/physiopathology
- Renal Insufficiency, Chronic/therapy
- Secretory Pathway/drug effects
- Wnt Proteins/metabolism
Collapse
Affiliation(s)
- Chien-Lin Lu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Jia-Fwu Shyu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan.
| | - Chia-Chao Wu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan.
| | - Chi-Feng Hung
- School of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan.
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu-Jen Catholic University, New Taipei City 262, Taiwan.
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan.
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan.
| | - Wen-Chih Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Tungs' Taichung MetroHarbor Hospital, Taichung City 433, Taiwan.
| | - Cai-Mei Zheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11103, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei City 235, Taiwan.
| | - Yi-Chou Hou
- Division of Nephrology, Department of Medicine, Cardinal-Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 23155, Taiwan.
| | - Yuh-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11103, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei City 235, Taiwan.
| | - Kuo-Cheng Lu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| |
Collapse
|
32
|
Wang FS, Wu RW, Lain WS, Tsai TC, Chen YS, Sun YC, Ke HJ, Li JC, Hwang J, Ko JY. Sclerostin vaccination mitigates estrogen deficiency induction of bone mass loss and microstructure deterioration. Bone 2018; 112:24-34. [PMID: 29653294 DOI: 10.1016/j.bone.2018.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 03/19/2018] [Accepted: 04/09/2018] [Indexed: 12/21/2022]
Abstract
Sclerostin (SOST) is a Wnt signaling inhibitor detrimental to osteogenic differentiation and bone mineral acquisition. While control of SOST action delays the pathogenesis of skeletal disorders, the effects of SOST vaccination on the estrogen deficiency-induced bone deterioration remain elusive. In this study, we generated a SOST-Fc fusion protein which was composed of a SOST peptide Pro-Asn-Ala-Ile-Gly along with an IgG Fc fragment. SOST-Fc vaccination increased serum anti-SOST antibody levels and reduced serum SOST concentrations in mice. In vitro, anti-SOST serum attenuated the SOST-induced inhibition of osteogenic gene expression in osteoblast cultures. Administration with SOST-Fc increased serum levels of bone formation marker osteocalcin and alleviated the ovariectomy escalation of serum resorption markers CTX-1 and TRAP5b concentrations. It remarkably lessened the estrogen deficiency-mediated deterioration of bone mineral density, morphometric characteristics of trabecular bone, and mechanical strength of femurs and lumbar spines. The SOST-Fc-treated skeletal tissue exhibited moderate responses to the adverse actions of ovariectomy to bone mineral accretion, osteoclast surface, trabecular separation, and fatty marrow histopathology. SOST-Fc treatment increased serum osteoclast-inhibitory factor osteoprotegrin levels in conjunction with strong Wnt3a, β-catenin, and TCF4 immunostaining in osteoblasts, whereas it weakened the estrogen deficiency enhancement of osteoclast-promoting factor receptor activator of nuclear factor-κB ligand. Taken together, blockade of SOST action by SOST-Fc vaccination sustains Wnt signaling, which harmonizes bone mineral accretion and resorption reactions and thereby ameliorates ovariectomy-induced bone loss. This study highlights SOST-Fc fusion protein as a new molecular therapeutic potential for preventing from osteoporotic disorders.
Collapse
Affiliation(s)
- Feng-Sheng Wang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Core Laboratory for Phenomics and Diagonistics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| | - Re-Wen Wu
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Wei-Shiung Lain
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Core Laboratory for Phenomics and Diagonistics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Tsai-Chen Tsai
- Core Laboratory for Phenomics and Diagonistics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Shan Chen
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Core Laboratory for Phenomics and Diagonistics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yi-Chih Sun
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Core Laboratory for Phenomics and Diagonistics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Huei-Jing Ke
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Core Laboratory for Phenomics and Diagonistics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Jui-Chen Li
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Core Laboratory for Phenomics and Diagonistics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Jaulang Hwang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Jih-Yang Ko
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
33
|
Drake MT, Fenske JS, Blocki FA, Zierold C, Appelman-Dijkstra N, Papapoulos S, Khosla S. Validation of a novel, rapid, high precision sclerostin assay not confounded by sclerostin fragments. Bone 2018; 111:36-43. [PMID: 29550267 PMCID: PMC5924723 DOI: 10.1016/j.bone.2018.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/13/2018] [Accepted: 03/13/2018] [Indexed: 10/17/2022]
Abstract
Sclerostin is a 190 amino acid protein secreted primarily by osteocytes. It was initially identified due to mutations in the SOST gene associated with high bone mass phenotypes. Much recent work has sought to determine the importance of sclerostin across an array of conditions which affect the human skeleton. However, accurate measurement of sclerostin from serum and plasma sources remains a significant impediment, with currently available commercial assays showing marked differences in measured sclerostin values. Accordingly, sclerostin assay standardization remains an important but unmet need before sclerostin measurements can be used for the clinical management of bone disease. Here we characterize a novel automated chemiluminescent sclerostin assay (LIAISON®, DiaSorin) which overcomes many of these limitations. Important assay characteristics include: a wide dynamic range (50-6500pg/mL); high intra- (<2.5%) and inter- (<5%) assay precision; matched serum and plasma equivalence (<10% difference); specificity for the intact sclerostin molecule; and rapid assay results. Serum sclerostin levels measured with the LIAISON® assay in a population-based sample of adult men (n=278) and women (n=348) demonstrated that sclerostin levels were significantly higher in men as compared to women and were positively associated with age in both sexes, consistent with previously published work. In postmenopausal women, serum sclerostin levels measured with the LIAISON® assay were reduced in response to treatment with either estrogen or teriparatide, again consistent with previous findings. Collectively, the above data demonstrate that the LIAISON® sclerostin assay provides a reliable tool for more confident assessment of emergent mechanisms wherein sclerostin may impact a number of bone related pathologies.
Collapse
Affiliation(s)
- Matthew T Drake
- Department of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | | | - Frank A Blocki
- DiaSorin Inc., 1951 Northwestern Ave, Stillwater, MN, USA
| | | | | | - Socrates Papapoulos
- Center for Bone Quality, Leiden University Medical Center, Leiden, Netherlands
| | - Sundeep Khosla
- Department of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
34
|
Hoseth EZ, Krull F, Dieset I, Mørch RH, Hope S, Gardsjord ES, Steen NE, Melle I, Brattbakk HR, Steen VM, Aukrust P, Djurovic S, Andreassen OA, Ueland T. Exploring the Wnt signaling pathway in schizophrenia and bipolar disorder. Transl Psychiatry 2018; 8:55. [PMID: 29507296 PMCID: PMC5838215 DOI: 10.1038/s41398-018-0102-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/28/2017] [Accepted: 12/05/2017] [Indexed: 01/05/2023] Open
Abstract
The Wnt signaling pathway plays a crucial role in neurodevelopment and in regulating the function and structure of the adult nervous system. Schizophrenia (SCZ) and bipolar disorder (BD) are severe mental disorders with evidence of subtle neurodevelopmental, structural and functional neuronal abnormalities. We aimed to elucidate the role of aberrant regulation of the Wnt system in these disorders by evaluating plasma levels of secreted Wnt modulators in patients (SCZ = 551 and BD = 246) and healthy controls (HCs = 639) using enzyme immune-assay. We also investigated the expression of 141 Wnt-related genes in whole blood in a subsample (SCZ = 338, BD = 241, and HCs = 263) using microarray analysis. Both SCZ and BD had dysregulated mRNA expression of Wnt-related genes favoring attenuated canonical (beta-catenin-dependent) signaling, and there were also indices of enhanced non-canonical Wnt signaling. In particular, FZD7, which may activate all Wnt pathways, but favors non-canonical signaling, and NFATc3, a downstream transcription factor and readout of the non-canonical Wnt/Ca2+ pathway, were significantly increased in SCZ and BD (p < 3 × 10-4). Furthermore, patients had lower plasma levels of soluble dickkopf 1 and sclerostin (p < 0.01) compared with HC. Our findings suggest that SCZ and BD are characterized by abnormal Wnt gene expression and plasma protein levels, and we propose that drugs targeting the Wnt pathway may have a role in the treatment of severe mental disorders.
Collapse
Affiliation(s)
- Eva Z. Hoseth
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway ,Division of Mental Health and Addiction, Møre and Romsdal Hospital Trust, Kristiansund, Norway
| | - Florian Krull
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ingrid Dieset
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ragni H. Mørch
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Sigrun Hope
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway ,0000 0004 0389 8485grid.55325.34Departent of Neurohabilitation, Division of Neurology, Oslo University Hospital, Oslo, Norway
| | - Erlend S. Gardsjord
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Nils Eiel Steen
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ingrid Melle
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Hans-Richard Brattbakk
- 0000 0004 1936 7443grid.7914.bNORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Oslo, Norway ,0000 0000 9753 1393grid.412008.fDr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Oslo, Norway
| | - Vidar M. Steen
- 0000 0004 1936 7443grid.7914.bNORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Oslo, Norway ,0000 0000 9753 1393grid.412008.fDr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Oslo, Norway
| | - Pål Aukrust
- 0000 0004 0389 8485grid.55325.34Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway ,0000 0004 0389 8485grid.55325.34Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway ,0000 0004 0389 8485grid.55325.34Instiute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway ,0000 0004 1936 8921grid.5510.1K.G. Jensen Inflammatory Research Center, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- 0000 0004 0389 8485grid.55325.34Department of Medical Genetics, Oslo University Hospital, Oslo, Norway ,0000 0004 1936 7443grid.7914.bNORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ole A. Andreassen
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway. .,Instiute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway. .,K.G. Jensen Inflammatory Research Center, University of Oslo, Oslo, Norway. .,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway.
| |
Collapse
|
35
|
Zeng C, Guo C, Cai J, Tang C, Dong Z. Serum sclerostin in vascular calcification and clinical outcome in chronic kidney disease. Diab Vasc Dis Res 2018; 15:99-105. [PMID: 29168393 DOI: 10.1177/1479164117742316] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Sclerostin, a potent soluble inhibitor of the Wnt signalling pathway, is known to inhibit bone formation by suppressing osteocytes differentiation and function. Patients with chronic kidney disease have high levels of serum sclerostin. Sclerostin has been implicated in the pathogenesis of vascular calcification, which may promote the cardiovascular events of morbidity and mortality in chronic kidney disease patients. However, the role of sclerostin in vascular calcification and clinical prognosis in chronic kidney disease remains elusive. While some studies suggested a positive correlation between serum sclerostin and vascular calcification or clinical outcome, other studies showed no or even negative correlation between them. Small sample size, heterogeneity in enrolled patients, discrepancy in anatomical structure examined and differences in the applied assays may be responsible for the discrepant results. Nonetheless, anti-sclerostin antibodies may be a new therapeutic approach to increase bone mass and strength in chronic kidney disease. This review aims to have a better understanding of the relationship of serum sclerostin with vascular calcification and clinical outcome in chronic kidney disease patients, and propose the application of anti-sclerostin therapy in chronic kidney disease.
Collapse
Affiliation(s)
- Cong Zeng
- 1 Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chunyuan Guo
- 2 Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Juan Cai
- 1 Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chengyuan Tang
- 1 Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zheng Dong
- 1 Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- 2 Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| |
Collapse
|
36
|
Disthabanchong S, Vipattawat K, Phakdeekitcharoen B, Kitiyakara C, Sumethkul V. Abdominal aorta and pelvic artery calcifications on plain radiographs may predict mortality in chronic kidney disease, hemodialysis and renal transplantation. Int Urol Nephrol 2018; 50:355-364. [PMID: 29236239 DOI: 10.1007/s11255-017-1758-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 11/20/2017] [Indexed: 02/05/2023]
Abstract
PURPOSE Vascular calcification is common in chronic kidney disease (CKD) and predicts poor patient outcomes. While computed tomography is the gold standard for evaluation of vascular calcification, plain radiograph offers a simpler and less costly alternative. The calcification of abdominal aorta, iliac and femoral arteries has been evaluated by plain radiograph, but the data on their outcome predictabilities are still limited. The present study investigated the role of abdominal aortic calcification (AAC) and pelvic arterial calcification (PAC) in predicting overall morality in non-dialysis CKD stages 2-5 (CKD 2-5), maintenance hemodialysis (HD) and long-term kidney transplant (KT) patients. METHODS Four hundred and nineteen patients were included. Lateral abdominal and pelvic radiographs were obtained. The degree of AAC and PAC was evaluated according to the methods described previously by Kaupplia et al. and Adragao et al. Patients were followed prospectively for 5 years. RESULTS AAC and PAC scores correlated well with the correlation coefficients of 0.442 for CKD 2-5, 0.438 for HD and 0.586 for KT (p < 0.001). Patients with AAC score > 6 or PAC score > 1 were older, showed higher prevalence of DM and had higher serum phosphate and PTH but lower serum albumin and eGFR. A more severe degree of AAC was associated with an increase in KT duration, whereas a more severe degree of PAC was associated with worsening kidney function and prolonged dialysis vintage. Kaplan-Meier survival curves revealed AAC score > 6 as a significant predictor of all-cause mortality in CKD 2-5 but not in HD or KT, whereas PAC score > 1 was a significant predictor of all-cause mortality in all three populations. After adjusting for age, the predictability of AAC was lost, whereas PAC remained an independent predictor of mortality in all three populations. Adjustments for cardiovascular and CKD risk factors including age, gender, BMI, DM, serum albumin, calcium and phosphate attenuated the predictability of PAC in HD but not in CKD 2-5 or KT patients. CONCLUSION PAC was better than AAC in predicting mortality in CKD, HD and KT patients.
Collapse
Affiliation(s)
- Sinee Disthabanchong
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama VI Rd, 7th floor, Building 1, Phayathai, Bangkok, 10400, Thailand.
- Ramathibodi Organ Transplant Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| | - Kotcharat Vipattawat
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama VI Rd, 7th floor, Building 1, Phayathai, Bangkok, 10400, Thailand
| | - Bunyong Phakdeekitcharoen
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama VI Rd, 7th floor, Building 1, Phayathai, Bangkok, 10400, Thailand
- Ramathibodi Organ Transplant Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Chagriya Kitiyakara
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama VI Rd, 7th floor, Building 1, Phayathai, Bangkok, 10400, Thailand
- Ramathibodi Organ Transplant Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Vasant Sumethkul
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270 Rama VI Rd, 7th floor, Building 1, Phayathai, Bangkok, 10400, Thailand
- Ramathibodi Organ Transplant Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
37
|
Saag KG, Petersen J, Grauer A. Romosozumab versus Alendronate and Fracture Risk in Women with Osteoporosis. N Engl J Med 2018; 378:195. [PMID: 29320649 DOI: 10.1056/nejmc1714810] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
38
|
Bisson SK, Ung RV, Mac-Way F. Role of the Wnt/ β-Catenin Pathway in Renal Osteodystrophy. Int J Endocrinol 2018; 2018:5893514. [PMID: 29808090 PMCID: PMC5901476 DOI: 10.1155/2018/5893514] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/08/2018] [Indexed: 12/17/2022] Open
Abstract
Vascular calcification and bone fragility are common and interrelated health problems that affect chronic kidney disease (CKD) patients. Bone fragility, which leads to higher risk of fracture and mortality, arises from the abnormal bone remodeling and mineralization that are seen in chronic kidney disease. Recently, sclerostin and Dickkopf-related protein 1 were suggested to play a significant role in CKD-related bone disease as they are known inhibitors of the Wnt pathway, thus preventing bone formation. This review focuses on new knowledge about the Wnt pathway in bone, how its function is affected by chronic kidney disease and how this affects bone structure. Expression of components and inhibitors of the Wnt pathway has been shown to be affected by the loss of kidney function, and a better understanding of the bone effects of Wnt pathway inhibitors could allow the development of new therapies to prevent bone fragility in this population.
Collapse
Affiliation(s)
- Sarah-Kim Bisson
- Faculty and Department of Medicine, CHU de Québec Research Center, L'Hôtel-Dieu de Québec Hospital, Endocrinology and Nephrology Axis, Université Laval, Quebec, Canada
| | - Roth-Visal Ung
- Faculty and Department of Medicine, CHU de Québec Research Center, L'Hôtel-Dieu de Québec Hospital, Endocrinology and Nephrology Axis, Université Laval, Quebec, Canada
| | - Fabrice Mac-Way
- Faculty and Department of Medicine, CHU de Québec Research Center, L'Hôtel-Dieu de Québec Hospital, Endocrinology and Nephrology Axis, Université Laval, Quebec, Canada
| |
Collapse
|
39
|
Delanaye P, Paquot F, Bouquegneau A, Blocki F, Krzesinski JM, Evenepoel P, Pottel H, Cavalier E. Sclerostin and chronic kidney disease: the assay impacts what we (thought to) know. Nephrol Dial Transplant 2017; 33:1404-1410. [DOI: 10.1093/ndt/gfx282] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/23/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
- Pierre Delanaye
- Department of Nephrology, Dialysis, Hypertension, Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
| | - François Paquot
- Department of Nephrology, Dialysis, Hypertension, Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
| | - Antoine Bouquegneau
- Department of Nephrology, Dialysis, Hypertension, Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
| | - Frank Blocki
- Medical LIAISON Bone & Mineral, DiaSorin Inc, Stillwater, MN, USA
| | - Jean-Marie Krzesinski
- Department of Nephrology, Dialysis, Hypertension, Transplantation, University of Liège (ULg CHU), CHU Sart Tilman, Liège, Belgium
| | - Pieter Evenepoel
- Department of Nephrology and Renal Transplantation, University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Hans Pottel
- Department of Public Health and Primary Care at Kulak, University of Leuven, Kortrijk, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, University of Liège, CHU Sart Tilman, Liège, Belgium
| |
Collapse
|
40
|
Wyatt CM, Drueke TB. Vascular calcification in chronic kidney disease: here to stay? Kidney Int 2017; 92:276-278. [DOI: 10.1016/j.kint.2017.05.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 05/15/2017] [Indexed: 11/25/2022]
|
41
|
Tartaglione L, Pasquali M, Rotondi S, Muci ML, Leonangeli C, Farcomeni A, Fassino V, Mazzaferro S. Interactions of sclerostin with FGF23, soluble klotho and vitamin D in renal transplantation. PLoS One 2017; 12:e0178637. [PMID: 28558021 PMCID: PMC5448809 DOI: 10.1371/journal.pone.0178637] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 05/16/2017] [Indexed: 01/05/2023] Open
Abstract
Relationships of Sclerostin, a bone anti-anabolic protein, with biomarkers of mineral bone disorders in chronic kidney disease are still unsettled, in particular in kidney transplant (KTR). In 80 KTR patients (31F/49M, 54.7±10.3 years) we studied the relationships of serum Sclerostin with eGFR, Calcium, Phosphate, Alkaline Phosphatase (AP), intact Parathyroid hormone (iPTH), soluble alpha-Klotho (sKlotho), intact Fibroblast Growth Factor 23 (iFGF23), 25-hydroxyvitamin D(25D) and 1,25-dihydroxyvitamin D (1,25D). Thirty healthy subjects (35.0±12.4 years, eGFR 109.1±14.1 ml /min/1,73m2) served as control for Sclerostin, iFGF23 and sKlotho. With a median eGFR of 46.3 mL/min/1.73m2 (IQR, 36.2–58.3) our KTR had median Sclerostin levels of 23.7 pmol/L (IQR: 20.8–32.8), not different from controls (26.6 pmol/L, IQR: 22.0–32.2; p = n.s). Sclerostin correlated negatively with AP (r = -.251; p = 0.023) and positively with iFGF23 (r = .227; p = 0.017) and 25D (r = .214; p = 0.025). Age-adjusted multiple regression analysis identified AP and 1,25D as negative and 25D and sKlotho as positive best predictors of Sclerostin. No correlation was evident with eGFR. The negative correlation with AP confirms the direct anti-anabolic role of Sclerostin. The associations either negative or positive with iFGF23, sKlotho, and vitamin D metabolites suggest also a modulatory role in mineral homeostasis. In particular, the associations with iFGF23 (positive) and 1,25D (negative) underline the relevant inhibitory action of Sclerostin on vitamin D activation. In conclusion, Sclerostin levels in KTR are normal and influenced more by bone turnover than by eGFR. Its involvement with other hormones of mineral homeostasis (FGF23/Klotho and Vitamin D) is part of the sophisticated cross-talk between bone and the kidney.
Collapse
Affiliation(s)
- Lida Tartaglione
- Department of Cardiovascular, Respiratory, Nephrologic, Anesthesiologic and Geriatric Sciences, Sapienza University of Rome, Italy
| | - Marzia Pasquali
- Department of Nephrology and Dialysis, University Hospital Company, Policlinico Umberto I, Rome, Italy
| | - Silverio Rotondi
- Department of Cardiovascular, Respiratory, Nephrologic, Anesthesiologic and Geriatric Sciences, Sapienza University of Rome, Italy
| | - Maria Luisa Muci
- Department of Cardiovascular, Respiratory, Nephrologic, Anesthesiologic and Geriatric Sciences, Sapienza University of Rome, Italy
| | - Cristiana Leonangeli
- Department of Cardiovascular, Respiratory, Nephrologic, Anesthesiologic and Geriatric Sciences, Sapienza University of Rome, Italy
| | - Alessio Farcomeni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy
| | - Valeria Fassino
- Department of Internal Medicine and medical Specialties, University Hospital Company, Policlinico Umberto I, Rome, Italy
| | - Sandro Mazzaferro
- Department of Cardiovascular, Respiratory, Nephrologic, Anesthesiologic and Geriatric Sciences, Sapienza University of Rome, Italy
- * E-mail:
| |
Collapse
|
42
|
Zhou H, Yang M, Li M, Cui L. Radial artery sclerostin expression in chronic kidney disease stage 5 predialysis patients: a cross-sectional observational study. Int Urol Nephrol 2017; 49:1433-1437. [PMID: 28455660 DOI: 10.1007/s11255-017-1604-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 04/20/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Bone metabolism disorder is often associated with cardiovascular calcification in patients with chronic kidney disease (CKD). Sclerostin, a novel candidate protein, has been identified to be involved in the bone-vascular axis. The aims of the current investigation were to assess vessel sclerostin expression and its relationship with circulating sclerostin levels. METHODS A cross-sectional observational study was conducted from January 2012 to December 2014. Thirty-two predialysis patients with CKD stage 5 who received arteriovenous fistula (AVF) operations were enrolled in this study. Radial arteries were collected and paraffin-embedded during the AVF operation, followed by immunohistochemical staining for sclerostin expression. In addition, serum sclerostin levels were measured by the enzyme-linked immunosorbent assay. RESULTS The prevalence of positive sclerostin staining in the radial arteries was 56.25%. Sclerostin expression was localized in the artery media layer. Serum sclerostin levels in patients with positive sclerostin expression were much higher than in those with negative expression (p = 0.018). Multivariate logistic regression analyses including potential confounders as age, gender, systolic blood pressure (BP), diastolic BP, serum sclerostin, corrected calcium (Ca), phosphate (P), Ca × P product, alkaline phosphatase, intact parathyroid hormone, and estimated glomerular filtration rate showed that only serum sclerostin levels were closely related to vessel sclerostin expression (p = 0.025). The area under the curve of serum sclerostin levels for predicting positive vessel sclerostin expression was 0.742 with 61.1% sensitivity and 85.7% specificity (p = 0.008). The cutoff point for vessel sclerostin expression of serum sclerostin was 1591.53 pg/mL. CONCLUSIONS Positive expression of sclerostin in the radial artery media layer was related to high serum sclerostin levels. Sclerostin may act as both a local and systemic regulator involved in vascular calcification.
Collapse
Affiliation(s)
- Hua Zhou
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Min Yang
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Min Li
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Li Cui
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China.
| |
Collapse
|
43
|
|
44
|
Weivoda MM, Youssef SJ, Oursler MJ. Sclerostin expression and functions beyond the osteocyte. Bone 2017; 96:45-50. [PMID: 27888056 PMCID: PMC5328839 DOI: 10.1016/j.bone.2016.11.024] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 11/17/2016] [Accepted: 11/21/2016] [Indexed: 11/24/2022]
Abstract
Sclerostin, the product of the SOST gene, is a secreted inhibitor of Wnt signaling that is produced by osteocytes to regulate bone formation. While it is often considered an osteocyte-specific protein, SOST expression has been reported in numerous other cell types, including hypertrophic chondrocytes and cementocytes. Of interest, SOST/sclerostin expression is altered in certain pathogenic conditions, including osteoarthritis and rheumatic joint disease, and it is unclear whether sclerostin plays a protective role or whether sclerostin may mediate disease pathogenesis. Therefore, as anti-sclerostin antibodies are being developed for the treatment of osteoporosis, it is important to understand the functions of sclerostin beyond the regulation of bone formation.
Collapse
Affiliation(s)
- Megan M Weivoda
- Division of Endocrinology, Metabolism, Nutrition & Diabetes, Mayo Clinic, USA.
| | - Stephanie J Youssef
- Division of Endocrinology, Metabolism, Nutrition & Diabetes, Mayo Clinic, USA
| | - Merry Jo Oursler
- Division of Endocrinology, Metabolism, Nutrition & Diabetes, Mayo Clinic, USA
| |
Collapse
|
45
|
|
46
|
Touw WA, Ueland T, Bollerslev J, Schousboe JT, Lim WH, Wong G, Thompson PL, Kiel DP, Prince RL, Rivadeneira F, Lewis JR. Association of Circulating Wnt Antagonists With Severe Abdominal Aortic Calcification in Elderly Women. J Endocr Soc 2017; 1:26-38. [PMID: 29264443 PMCID: PMC5677217 DOI: 10.1210/js.2016-1040] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 12/16/2016] [Indexed: 12/22/2022] Open
Abstract
Context There is great interest in the biology of vascular calcification. Wnt/β-catenin signaling is an important mediator of mineralization and may play a role in vascular calcification. Objective We assessed the association between circulating Wnt antagonists and abdominal aortic calcification (AAC) severity in elderly women. Design This was a cross-sectional analysis of the Calcium Intake Fracture Outcome Study. Setting The participants were recruited from the community-dwelling elderly population. Participants We examined 768 women aged over 70 years. Interventions We collected blood samples, and lateral spine images captured during bone density assessment were used to score AAC with a validated 24-point scale. Main Outcome Measures We tested the hypothesis that low Wnt antagonist levels of Dickkopf-1 (DKK1), secreted frizzled related protein 3 (sFRP3), and Wnt inhibitory factor 1 (WIF1) are associated with severe AAC (AAC24 score > 5). Results Severe AAC was present in 146 women (19%). Lower levels of DKK1, but not WIF1 and sFRP3, were associated with higher odds of severe AAC. Per standard deviation decrease in DKK1 was associated with increased multivariable-adjusted odds ratio (OR) of severe AAC [OR, 1.26; 95% confidence interval (CI), 1.04 to 1.52; P = 0.017]. In quartile analyses, the lowest and second-lowest quartiles of DKK1 had increased multivariable-adjusted odds of severe AAC vs the highest quartile (OR, 2.05; 95% CI, 1.18 to 3.56; P = 0.011 and OR, 1.83; 95% CI, 1.05 to 3.19; P = 0.035). Conclusions In elderly women, DKK1, but not sFRP3 or WIF1, is associated with severe AAC. This study supports the concept that Wnt/β-catenin signaling is an important regulator of vascular mineral metabolism and is independent of other risk factors.
Collapse
Affiliation(s)
- Wilhelmina A Touw
- Erasmus University, Rotterdam 3015GE, The Netherlands.,University of Western Australia School of Medicine and Pharmacology
| | | | - Jens Bollerslev
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Rikshospitalet, N-0514 Oslo, Norway.,University of Oslo, N-0315 Oslo, Norway
| | - John T Schousboe
- Park Nicollet Osteoporosis Center and HealthPartners Institute, Minneapolis, Minnesota 55416.,Division of Health Policy and Management, University of Minnesota, Minneapolis, Minnesota 55455
| | - Wai H Lim
- University of Western Australia School of Medicine and Pharmacology.,Department of Renal Medicine
| | - Germaine Wong
- Centre for Kidney Research, Children's Hospital at Westmead, School of Public Health, Sydney Medical School, The University of Sydney, Sydney, 2145 Australia
| | | | - Douglas P Kiel
- Institute for Aging Research, Hebrew Senior Life, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02131; and
| | - Richard L Prince
- University of Western Australia School of Medicine and Pharmacology.,Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, 6009 Australia
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam 3015GE, The Netherlands
| | - Joshua R Lewis
- University of Western Australia School of Medicine and Pharmacology.,Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, 6009 Australia.,Centre for Kidney Research, Children's Hospital at Westmead, School of Public Health, Sydney Medical School, The University of Sydney, Sydney, 2145 Australia
| |
Collapse
|
47
|
Jankowska M, Haarhaus M, Qureshi AR, Lindholm B, Evenepoel P, Stenvinkel P. Sclerostin─A Debutant on the Autosomal Dominant Polycystic Kidney Disease Scene? Kidney Int Rep 2017; 2:481-485. [PMID: 29142975 PMCID: PMC5678633 DOI: 10.1016/j.ekir.2017.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 12/09/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022] Open
Abstract
Introduction Autosomal dominant polycystic kidney disease (ADPKD) is a genetic disease originating from a mutation in genes encoding polycystin 1 and 2. Recent evidence suggests that these polycystins mediate mechanosensation not only in the primary cilium of kidney cells but also in bone cells. The Wnt/β-catenin signaling pathway plays a central role in mechanotransduction in osteocytes. Mechanical unloading causes the upregulation of the Wnt inhibitor sclerostin. We tested the hypothesis that ADPKD associates with higher circulating sclerostin levels. Methods In this observational, cross-sectional study, circulating levels of sclerostin and other laboratory parameters of mineral and bone disease, including intact parathyroid hormone (PTH), calcium, phosphate, magnesium, 25(OH) D-vitamin, 1,25 (OH)2 D-vitamin, and bone specific alkaline phosphatase (BALP) were assessed in 100 patients with end-stage renal disease recruited from an ongoing longitudinal cohort study in Stockholm, Sweden. Results Patients with ADPKD had higher sclerostin levels and lower BALP levels as compared to patients with other primary renal disease. In multivariate analysis, ADPKD associated with circulating sclerostin levels, independent of the established determinants including age, gender, body mass index, diabetes, phosphate, PTH, and 1,25 (OH)2 D-vitamin. Discussion Circulating sclerostin levels are increased in ADPKD, possibly reflecting impaired mechanosensation. The clinical relevance of this finding, especially with regard to bone health, remains to be investigated. Our finding draws attention to the etiology of kidney disease as an important, yet neglected, confounder of the association between renal failure and mineral and bone disease.
Collapse
Affiliation(s)
- Magdalena Jankowska
- Division of Renal Medicine and Baxter Novum, Karolinska University Hospital at Huddinge, Karolinska Institutet, Stockholm, Sweden.,Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Mathias Haarhaus
- Division of Renal Medicine and Baxter Novum, Karolinska University Hospital at Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Abdul Rashid Qureshi
- Division of Renal Medicine and Baxter Novum, Karolinska University Hospital at Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Lindholm
- Division of Renal Medicine and Baxter Novum, Karolinska University Hospital at Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Pieter Evenepoel
- Department of Immunology and Microbiology, Laboratory of Nephrology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Karolinska University Hospital at Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
48
|
Kanbay M, Solak Y, Siriopol D, Aslan G, Afsar B, Yazici D, Covic A. Sclerostin, cardiovascular disease and mortality: a systematic review and meta-analysis. Int Urol Nephrol 2016; 48:2029-2042. [DOI: 10.1007/s11255-016-1387-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/27/2016] [Indexed: 12/23/2022]
|
49
|
NasrAllah MM, Nassef A, Elshaboni TH, Morise F, Osman NA, Sharaf El Din UA. Comparing different calcification scores to detect outcomes in chronic kidney disease patients with vascular calcification. Int J Cardiol 2016; 220:884-9. [PMID: 27400189 DOI: 10.1016/j.ijcard.2016.06.064] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/19/2016] [Indexed: 02/09/2023]
Abstract
BACKGROUND There is no consensus on the most appropriate technique to diagnose vascular calcification in chronic kidney disease. This is primarily because of the absence of direct comparisons of predictive values of the various calcification scores, especially outside the coronary vascular beds, to detect clinical outcomes. METHODS We included 93 haemodialysis patients and performed 6 vascular calcification scores: two scores utilised simple X-rays of abdominal aorta and peripheral vessels. CT scans of the thoracic, upper abdominal and lower abdominal aorta were performed to calculate the aortic calcification index and CT of the pelvis for calcification of iliac vessels. Patients were followed for 63months (mean 46.8months) for first major cardiovascular events and mortality. RESULTS Nineteen cardiovascular events and 28 deaths occurred. Calcification was detected more sensitively in central and peripheral beds using CT scans compared to X-rays (p<0.001). CT scans detected calcification more frequently in distal than proximal vascular beds (p<0.001). Calcification of the pelvic vessels and lower abdominal aorta were most predictive of events including pre-existing cardiovascular disease O.R. 6.5 (95% C.I. 2-22; p=0.001) and O.R. 3 (95% C.I. 1.1-9; p=0.035); new major cardiovascular events H.R. 4.2 (95% C.I. 1.5-11; p=0.006) and H.R. 2 (95% C.I. 0.8-5.3; p=0.1) as well as mortality H.R. 2.8 (95% C.I. 1.3-6; p=0.01) and H.R. 2.2 (95% C.I. 1.04-5; p=0.04) respectively. CONCLUSIONS CT based techniques are more sensitive than plain X-rays at detecting peripheral and aortic vascular calcifications. Distal CT scans of the aorta and pelvic vessels have the highest predictive value for cardiovascular events and mortality.
Collapse
Affiliation(s)
- Mohamed M NasrAllah
- Kasr AlAiny School of Medicine, Cairo University, Nephrology, Saray Street, Manial, Cairo, Egypt.
| | - Amr Nassef
- Kasr AlAiny School of Medicine, Cairo University, Radiology, Cairo, Egypt
| | - Tarik H Elshaboni
- Kasr AlAiny School of Medicine, Cairo University, Nephrology, Saray Street, Manial, Cairo, Egypt
| | - Fadia Morise
- Kasr AlAiny School of Medicine, Cairo University, Nephrology, Saray Street, Manial, Cairo, Egypt
| | - Noha A Osman
- Kasr AlAiny School of Medicine, Cairo University, Nephrology, Saray Street, Manial, Cairo, Egypt
| | - Usama A Sharaf El Din
- Kasr AlAiny School of Medicine, Cairo University, Nephrology, Saray Street, Manial, Cairo, Egypt
| |
Collapse
|