1
|
Celebi Torabfam G, Porsuk MH. The Role of the Receptor Activator of Nuclear Factor Kappa-B Ligand/Osteoprotegerin Ratio in Vascular Diseases: A Therapeutic Approach. Angiology 2025; 76:309-322. [PMID: 38171493 DOI: 10.1177/00033197231226275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Cardiovascular and bone diseases contribute independently to mortality and global health. The exact mechanisms involved in the pathophysiology shared between bone and vascular diseases are not well defined. Endothelial cells and osteoblasts communicate during osteogenesis, thus establishing a connection between angiogenesis and osteogenesis. One shared mechanism may involve osteoprotegerin (OPG) and its ligand Receptor Activator of NF-κB Ligand (RANKL). The RANKL/OPG ratio is an important modulator for the skeletal, immunological, and vascular systems. OPG levels are elevated due to either osteogenic causes or inflammatory responses in the vasculature. The data obtained from clinical and in vitro studies support the role of the RANKL/OPG ratio as a potential marker for the progression of endothelial damage. Therefore, determining the therapeutic approaches for the targeting RANKL/OPG ratio and evaluating its usage as a biomarker in cardiovascular and bone pathophysiology are needed. By integrating the protective and disease-causing role of OPG with its ligand, this review outlines the role of the RANKL/OPG ratio at the molecular level. We also consider targeted therapeutic approaches.
Collapse
Affiliation(s)
- Gizem Celebi Torabfam
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, Istanbul, Turkey
| | - Melis Hazal Porsuk
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, Istanbul, Turkey
| |
Collapse
|
2
|
Low S, Pek S, Moh A, Liu JJ, Pandian B, Ang K, Tang WE, Lim Z, Subramaniam T, Sum CF, Lim SC. The association between osteoprotegerin and arterial stiffness in a 10-year longitudinal study of patients with type 2 diabetes. Diab Vasc Dis Res 2024; 21:14791641241304435. [PMID: 39626773 PMCID: PMC11615981 DOI: 10.1177/14791641241304435] [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: 06/27/2024] [Revised: 09/30/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
Introduction: Osteoprotegerin (OPG) inhibits vascular calcification which is central to pathogenesis of arterial stiffness. However, it promotes inflammation by upregulating expression of vascular cell adhesion molecule-1(VCAM-1), thereby contributing to arterial stiffness. We investigated longitudinal association between OPG and arterial stiffness in type 2 diabetes (T2D), causality of the association and mediation by VCAM-1. Methods: This was a prospective cohort study of T2D patients (N = 1877, mean age 57.0 ± 10.8) with 10 years' follow-up. Baseline plasma OPG was measured using immunoassay. Pulse wave velocity (PWV) was assessed using applanation tonometry. We examined association between OPG and follow-up PWV using linear mixed model. One-sample Mendelian Randomization (MR) was conducted with rs1385492 as OPG-associated single nucleotide polymorphism (SNP). Results: Baseline natural log (Ln)-transformed OPG was positively associated with baseline and follow-up PWV with adjusted coefficients 0.43 (95%CI 0.05, 0.80; p = .026) and 0.51 (95%CI 0.06 to 0.97; p = .028) respectively. Genetically-predicted higher levels of plasma OPG was associated with higher last follow-up PWV with coefficient 10.81 (95%CI 2.97, 18.65; p = .007) per unit increase in LnOPG. Higher VCAM-1 accounted for 10.2% of association between LnOPG and follow-up PWV. Discussion: Baseline plasma OPG was associated with higher follow-up PWV in patients with T2D, with genetic evidence from MR. This association may be mediated, at least in part, by VCAM-1.
Collapse
Affiliation(s)
- Serena Low
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
- Diabetes Centre, Admiralty Medical Centre, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Sharon Pek
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Angela Moh
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Jian-Jun Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
| | | | - Keven Ang
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
| | - Wern Ee Tang
- National Healthcare Group Polyclinics, Singapore, Singapore
| | - Ziliang Lim
- National Healthcare Group Polyclinics, Singapore, Singapore
| | | | - Chee Fang Sum
- Diabetes Centre, Admiralty Medical Centre, Singapore, Singapore
| | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore, Singapore
- Diabetes Centre, Admiralty Medical Centre, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| |
Collapse
|
3
|
Nair AS, Woodford J, Loughland J, Andrew D, Piera K, Amante F, William T, Grigg MJ, McCarthy JS, Anstey NM, Boyle MJ, Barber BE. Osteoprotegerin (OPG) and its ligands RANKL and TRAIL in falciparum, vivax and knowlesi malaria: correlations with disease severity, and B cell production of OPG. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.22.24310838. [PMID: 39108527 PMCID: PMC11302609 DOI: 10.1101/2024.07.22.24310838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Osteoprotegerin (OPG) is a soluble decoy receptor for receptor activator of NF-ƙB ligand (RANKL) and TNF-related apoptosis-inducing ligand (TRAIL), and is increasingly recognised as a marker of poor prognosis in a number of diseases. Here we demonstrate that in Malaysian adults with falciparum and vivax malaria, OPG is increased, and its ligands TRAIL and RANKL decreased, in proportion to disease severity. In volunteers experimentally infected with P. falciparum and P. vivax, RANKL was suppressed, while TRAIL was unexpectedly increased, suggesting binding of OPG to RANKL prior to TRAIL. We also demonstrate that P. falciparum stimulates B cells to produce OPG in vitro, and that B cell OPG production is increased ex vivo in patients with falciparum, vivax and knowlesi malaria. Our findings provide further evidence of the importance of the OPG/RANKL/TRAIL pathway in pathogenesis of diseases involving systemic inflammation, and may have implications for adjunctive therapies. Further evaluation of the role of B cell production of OPG in host responses to malaria and other inflammatory diseases is warranted.
Collapse
Affiliation(s)
| | - John Woodford
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, Bethesda, United States
| | - Jessica Loughland
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Burnet Institute, Melbourne, Australia
| | - Dean Andrew
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kim Piera
- Menzies School of Health Research, Darwin, Australia
| | - Fiona Amante
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | | | - James S McCarthy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | | - Michelle J Boyle
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Burnet Institute, Melbourne, Australia
| | - Bridget E Barber
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Menzies School of Health Research, Darwin, Australia
| |
Collapse
|
4
|
Dutka M, Garczorz W, Kosowska A, Buczek E, Godek P, Wojakowski W, Francuz T. Osteoprotegerin Is Essential for the Development of Endothelial Dysfunction Induced by Angiotensin II in Mice. Int J Mol Sci 2024; 25:6434. [PMID: 38928140 PMCID: PMC11203749 DOI: 10.3390/ijms25126434] [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: 05/11/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Opinions on the effects of osteoprotegerin (OPG) have evolved over the years from a protein protecting the vasculature from calcification to a cardiovascular risk factor contributing to inflammation within the vascular wall. Nowadays, the link between OPG and angiotensin II (Ang II) appears to be particularly important. In this study, the endothelial function was investigated in OPG-knockout mice (B6.129.S4-OPG, OPG-) and wild-type (C57BL/6J, OPG+) mice under basic conditions and after Ang II exposure by assessing the endothelium-dependent diastolic response of aortic rings to acetylcholine in vitro. A further aim of the study was to compare the effect of Ang II on the expression of cytokines in the aortic wall of both groups of mice. Our study shows that rings from OPG- mice had their normal endothelial function preserved after incubation with Ang II, whereas those from OPG+ mice showed significant endothelial dysfunction. We conclude that the absence of OPG, although associated with a pro-inflammatory cytokine profile in the vascular wall, simultaneously protects against Ang II-induced increases in pro-inflammatory cytokines in the murine vascular wall. Our study also demonstrates that the absence of OPG can result in a decrease in the concentration of pro-inflammatory cytokines in the vascular wall after Ang II exposure. The presence of OPG is therefore crucial for the development of Ang II-induced inflammation in the vascular wall and for the development of Ang II-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, 43-309 Bielsko-Biala, Poland
| | - Wojciech Garczorz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (W.G.); (A.K.); (T.F.)
| | - Agnieszka Kosowska
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (W.G.); (A.K.); (T.F.)
| | - Elzbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 30-348 Krakow, Poland;
| | - Piotr Godek
- Department of Cardiology and Structural Heart Disease, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (P.G.); (W.W.)
| | - Wojciech Wojakowski
- Department of Cardiology and Structural Heart Disease, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (P.G.); (W.W.)
| | - Tomasz Francuz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (W.G.); (A.K.); (T.F.)
| |
Collapse
|
5
|
Jeon HJ, Ryu JH, Kim MG, Huh KH, Lee KW, Kim CD, Kang KP, Ro H, Han S, Yang J. Association of Serum Osteoprotegerin With Vascular Calcification, and Cardiovascular and Graft Outcomes in Kidney Transplant Patients: Results From the KNOW-KT. Transplantation 2024; 108:1239-1248. [PMID: 38291579 DOI: 10.1097/tp.0000000000004903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
BACKGROUND Vascular calcification and stiffness contribute to increased cardiovascular morbidity in patients with chronic kidney disease. This study investigated associations between serum osteoprotegerin (OPG) levels and vascular calcification or stiffness to assess cardiovascular and graft outcomes in kidney transplant patients. METHODS The KoreaN cohort study for Outcome in patients With Kidney Transplantation was a prospective multicenter cohort study. Serum OPG levels were measured at baseline and 3 y after transplantation in 1018 patients. Patients were classified into high and low OPG groups according to median serum OPG levels. The median follow-up duration was 93.5 mo. RESULTS The mean age was 45.8 ± 11.7 y and 62.9% were men. Patients with high OPG had significantly higher coronary artery calcium scores, abdominal aortic calcification scores, and brachial-ankle pulse wave velocities than those with lower OPG; these parameters remained significant for 5 y after transplantation. The 3-y OPG levels were lower than baseline values ( P < 0.001) and were positively correlated ( r = 0.42, P < 0.001). Multivariate Cox regression analysis showed that high OPG levels were significantly associated with posttransplant cardiovascular events ( P = 0.008) and death-censored graft loss ( P = 0.004). Similar findings regarding posttransplant cardiovascular events ( P = 0.012) and death-censored graft loss ( P = 0.037) were noted in patients with high OPG at the 3-y follow-up. Mediation analyses revealed that coronary artery calcium scores, abdominal aortic calcification scores, and brachial-ankle pulse wave velocities could act as mediators between serum OPG levels and posttransplant cardiovascular events. CONCLUSIONS Serum OPG concentration is associated with vascular calcification and stiffness and could be a significant risk factor for cardiovascular outcomes and graft loss in patients undergoing kidney transplantation.
Collapse
Affiliation(s)
- Hee Jung Jeon
- Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Jung-Hwa Ryu
- Department of Internal Medicine, Ewha Womans University Medical Center, Seoul, Republic of Korea
| | - Myung-Gyu Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyu Ha Huh
- Department of Transplantation Surgery, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea
| | - Kyo Won Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Chan-Duck Kim
- Department of Internal Medicine, Kyungpook National University School of Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Kyung Pyo Kang
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Han Ro
- Department of Internal Medicine, Gachon University, Gil Hospital, Incheon, Republic of Korea
| | - Seungyeup Han
- Department of Internal Medicine, Keimyung University, Dongsan Medical Center, Daegu, Republic of Korea
| | - Jaeseok Yang
- Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, Seoul, Republic of Korea
| |
Collapse
|
6
|
Suh SH, Oh TR, Choi HS, Kim CS, Bae EH, Ma SK, Oh KH, Lee KB, Jeong JC, Jung JY, Kim SW. Circulating osteoprotegerin levels and cardiovascular outcomes in patients with pre-dialysis chronic kidney disease: results from the KNOW-CKD study. Sci Rep 2024; 14:4136. [PMID: 38374135 PMCID: PMC10876961 DOI: 10.1038/s41598-024-54335-y] [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: 09/15/2023] [Accepted: 02/12/2024] [Indexed: 02/21/2024] Open
Abstract
While the relationship between circulating osteoprotegerin (OPG) and cardiovascular events is well-established in the general population, its association with cardiovascular risks in chronic kidney disease (CKD) patients remains less robust. This study hypothesized that elevated circulating OPG levels might be associated with an increased risk of major adverse cardiac events (MACE) in CKD patients, a total of 2,109 patients with CKD stages 1 through pre-dialysis 5 from the KNOW-CKD cohort were categorized into quartiles based on serum OPG levels. The primary outcome of the study was 3-point MACE, defined as a composite of nonfatal myocardial infarction, nonfatal stroke, or cardiac death. The median follow-up duration was 7.9 years. The cumulative incidence of 3-point MACE significantly varied across serum OPG levels in Kaplan-Meier curve analysis (P < 0.001, log-rank test), with the highest incidence observed in the 4th quartile. Cox regression analysis indicated that, relative to the 1st quartile, the risk of 3-point MACE was significantly higher in the 3rd (adjusted hazard ratio 2.901, 95% confidence interval 1.009 to 8.341) and the 4th quartiles (adjusted hazard ratio 4.347, 95% confidence interval 1.410 to 13.395). In conclusion, elevated circulating OPG levels are associated with adverse cardiovascular outcomes in pre-dialysis CKD patients.
Collapse
Affiliation(s)
- Sang Heon Suh
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Tae Ryom Oh
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Hong Sang Choi
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Chang Seong Kim
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Eun Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyu-Beck Lee
- Division of Nephrology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jong Cheol Jeong
- Division of Nephrology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ji Yong Jung
- Division of Nephrology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea.
| |
Collapse
|
7
|
Fogarty H, Ahmad A, Atiq F, Doherty D, Ward S, Karampini E, Rehill A, Leon G, Byrne C, Geoghegan R, Conroy H, Byrne M, Budde U, Schneppenheim S, Sheehan C, Ngwenya N, Baker RI, Preston RJS, Tuohy E, McMahon C, O’Donnell JS. VWF-ADAMTS13 axis dysfunction in children with sickle cell disease treated with hydroxycarbamide vs blood transfusion. Blood Adv 2023; 7:6974-6989. [PMID: 37773926 PMCID: PMC10690561 DOI: 10.1182/bloodadvances.2023010824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/17/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023] Open
Abstract
Previous studies have reported elevated von Willebrand factor (VWF) levels in patients with sickle cell disease (SCD) and demonstrated a key role for the VWF-ADAMTS13 axis in the pathobiology of SCD vaso-occlusion. Although blood transfusion is the gold standard for stroke prevention in SCD, the biological mechanisms underpinning its improved efficacy compared with hydroxycarbamide are not fully understood. We hypothesized that the improved efficacy of blood transfusion might relate to differences in VWF-ADAMTS13 axis dysfunction. In total, 180 children with a confirmed diagnosis of SCD (hemoglobin SS) on hydroxycarbamide (n = 96) or blood transfusion (n = 84) were included. Despite disease-modifying treatment, plasma VWF and VWF propeptide were elevated in a significant proportion of children with SCD (33% and 47%, respectively). Crucially, all VWF parameters were significantly higher in the hydroxycarbamide compared with the blood transfusion cohort (P < .05). Additionally, increased levels of other Weibel-Palade body-stored proteins, including factor VIII (FVIII), angiopoietin-2, and osteoprotegerin were observed, indicated ongoing endothelial cell activation. Children treated with hydroxycarbamide also had higher FVIII activity and enhanced thrombin generation compared with those in the blood transfusion cohort (P < .001). Finally, hemolysis markers strongly correlated with VWF levels (P < .001) and were significantly reduced in the blood transfusion cohort (P < .001). Cumulatively, to our knowledge, our findings demonstrate for the first time that despite treatment, ongoing dysfunction of the VWF-ADAMTS13 axis is present in a significant subgroup of pediatric patients with SCD, especially those treated with hydroxycarbamide.
Collapse
Affiliation(s)
- Helen Fogarty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Azaz Ahmad
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ferdows Atiq
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Dearbhla Doherty
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Soracha Ward
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ellie Karampini
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aisling Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Gemma Leon
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ciara Byrne
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Rosena Geoghegan
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Helena Conroy
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Mary Byrne
- National Coagulation Centre, St. James’s Hospital, Dublin, Ireland
| | - Ulrich Budde
- Department of Haemostaseology, MVZ Medilys Laborgesellschaft mbH, Hamburg, Germany
| | - Sonja Schneppenheim
- Department of Haemostaseology, MVZ Medilys Laborgesellschaft mbH, Hamburg, Germany
| | - Ciara Sheehan
- Department of Haematology, St. James’s Hospital, Dublin, Ireland
| | - Noel Ngwenya
- Department of Haematology, St. James’s Hospital, Dublin, Ireland
| | - Ross I. Baker
- Western Australia Centre for Thrombosis and Haemostasis, Perth Blood Institute, Murdoch University, Perth, WA, Australia
- Irish-Australian Blood Collaborative Network, Dublin, Ireland and Perth, Australia
| | - Roger J. S. Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - Emma Tuohy
- Department of Haematology, St. James’s Hospital, Dublin, Ireland
| | - Corrina McMahon
- Department of Haematology, Children’s Health Ireland at Crumlin, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
| | - James S. O’Donnell
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
- National Coagulation Centre, St. James’s Hospital, Dublin, Ireland
- Irish-Australian Blood Collaborative Network, Dublin, Ireland and Perth, Australia
| |
Collapse
|
8
|
Tang K, Zhang Y, Zhang C, Hu H, Zhuang R, Jin B, Zhang Y, Ma Y. Hantaan virus-induced elevation of plasma osteoprotegerin and its clinical implications in hemorrhagic fever with renal syndrome. Int J Infect Dis 2023; 126:14-21. [PMID: 36371012 DOI: 10.1016/j.ijid.2022.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 10/13/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022] Open
Abstract
OBJECTIVES The bleeding tendency is a hallmark of hemorrhagic fever with renal syndrome (HFRS) after Hantaan virus (HTNV) infection. Growing reports indicate the importance of osteoprotegerin (OPG) in vascular homeostasis, implying OPG might be involved in the pathogenesis of coagulopathy in patients with HFRS. METHODS Acute and convalescence plasmas of 32 patients with HFRS were collected. Enzyme-linked immunosorbent assays (ELISA) were used to detect plasma OPG levels and other parameters. The human umbilical vein endothelial cells were stimulated with HTNV and/or tumor necrosis factor-α (TNF-α) to explore the source of OPG. RESULTS Plasma OPG levels of patients with HFRS were elevated and correlated positively with the severity of HFRS and negatively with platelet counts. Abundant OPG was released from endothelial cells in response to TNF-α stimuli, along with HTNV infection, which was in accordance with the findings of positive correlations between plasma OPG and TNF-α or c-reactive protein. Importantly, plasma OPG levels correlated positively with activated partial thromboplastin time and the content of d-dimer. CONCLUSION These findings suggested that increased plasma OPG levels induced by HTNV might be an important factor for the severity of HFRS, and was likely involved in endothelium dysfunction and hemorrhagic disorder of HFRS, which might contribute to the pathogenesis of hemorrhage in HFRS.
Collapse
Affiliation(s)
- Kang Tang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Yusi Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Chunmei Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Haifeng Hu
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ran Zhuang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Boquan Jin
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Yun Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China.
| | - Ying Ma
- Department of Immunology, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
9
|
Zeller J, Cheung Tung Shing KS, Nero TL, McFadyen JD, Krippner G, Bogner B, Kreuzaler S, Kiefer J, Horner VK, Braig D, Danish H, Baratchi S, Fricke M, Wang X, Kather MG, Kammerer B, Woollard KJ, Sharma P, Morton CJ, Pietersz G, Parker MW, Peter K, Eisenhardt SU. A novel phosphocholine-mimetic inhibits a pro-inflammatory conformational change in C-reactive protein. EMBO Mol Med 2022; 15:e16236. [PMID: 36468184 PMCID: PMC9832874 DOI: 10.15252/emmm.202216236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/29/2022] [Accepted: 11/06/2022] [Indexed: 12/09/2022] Open
Abstract
C-reactive protein (CRP) is an early-stage acute phase protein and highly upregulated in response to inflammatory reactions. We recently identified a novel mechanism that leads to a conformational change from the native, functionally relatively inert, pentameric CRP (pCRP) structure to a pentameric CRP intermediate (pCRP*) and ultimately to the monomeric CRP (mCRP) form, both exhibiting highly pro-inflammatory effects. This transition in the inflammatory profile of CRP is mediated by binding of pCRP to activated/damaged cell membranes via exposed phosphocholine lipid head groups. We designed a tool compound as a low molecular weight CRP inhibitor using the structure of phosphocholine as a template. X-ray crystallography revealed specific binding to the phosphocholine binding pockets of pCRP. We provide in vitro and in vivo proof-of-concept data demonstrating that the low molecular weight tool compound inhibits CRP-driven exacerbation of local inflammatory responses, while potentially preserving pathogen-defense functions of CRP. The inhibition of the conformational change generating pro-inflammatory CRP isoforms via phosphocholine-mimicking compounds represents a promising, potentially broadly applicable anti-inflammatory therapy.
Collapse
Affiliation(s)
- Johannes Zeller
- Department of Plastic and Hand Surgery, University of Freiburg Medical CentreMedical Faculty of the University of FreiburgFreiburgGermany,Baker Heart and Diabetes InstituteMelbourneVic.Australia
| | - Karen S Cheung Tung Shing
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVic.Australia,Department of Cardiometabolic HealthThe University of MelbourneParkvilleVic.Australia
| | - Tracy L Nero
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVic.Australia,Department of Cardiometabolic HealthThe University of MelbourneParkvilleVic.Australia,ACRF Rational Drug Discovery CentreSt. Vincent's Institute of Medical ResearchFitzroyVic.Australia
| | - James D McFadyen
- Baker Heart and Diabetes InstituteMelbourneVic.Australia,Department of Cardiometabolic HealthThe University of MelbourneParkvilleVic.Australia
| | - Guy Krippner
- Baker Heart and Diabetes InstituteMelbourneVic.Australia
| | - Balázs Bogner
- Department of Plastic and Hand Surgery, University of Freiburg Medical CentreMedical Faculty of the University of FreiburgFreiburgGermany
| | - Sheena Kreuzaler
- Department of Plastic and Hand Surgery, University of Freiburg Medical CentreMedical Faculty of the University of FreiburgFreiburgGermany
| | - Jurij Kiefer
- Department of Plastic and Hand Surgery, University of Freiburg Medical CentreMedical Faculty of the University of FreiburgFreiburgGermany
| | - Verena K Horner
- Department of Plastic and Hand Surgery, University of Freiburg Medical CentreMedical Faculty of the University of FreiburgFreiburgGermany
| | - David Braig
- Department of Plastic and Hand Surgery, University of Freiburg Medical CentreMedical Faculty of the University of FreiburgFreiburgGermany
| | - Habiba Danish
- Baker Heart and Diabetes InstituteMelbourneVic.Australia,School of Health and Biomedical SciencesRMIT UniversityMelbourneVic.Australia
| | - Sara Baratchi
- School of Health and Biomedical SciencesRMIT UniversityMelbourneVic.Australia
| | - Mark Fricke
- Department of Plastic and Hand Surgery, University of Freiburg Medical CentreMedical Faculty of the University of FreiburgFreiburgGermany
| | - Xiaowei Wang
- Baker Heart and Diabetes InstituteMelbourneVic.Australia,Department of Cardiometabolic HealthThe University of MelbourneParkvilleVic.Australia
| | - Michel G Kather
- Centre for Integrative Signalling Analysis CISAUniversity of FreiburgFreiburgGermany
| | - Bernd Kammerer
- Centre for Integrative Signalling Analysis CISAUniversity of FreiburgFreiburgGermany
| | | | - Prerna Sharma
- Baker Heart and Diabetes InstituteMelbourneVic.Australia
| | - Craig J Morton
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVic.Australia,Department of Cardiometabolic HealthThe University of MelbourneParkvilleVic.Australia
| | - Geoffrey Pietersz
- Baker Heart and Diabetes InstituteMelbourneVic.Australia,Department of Cardiometabolic HealthThe University of MelbourneParkvilleVic.Australia
| | - Michael W Parker
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVic.Australia,Department of Cardiometabolic HealthThe University of MelbourneParkvilleVic.Australia,ACRF Rational Drug Discovery CentreSt. Vincent's Institute of Medical ResearchFitzroyVic.Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes InstituteMelbourneVic.Australia,Department of Cardiometabolic HealthThe University of MelbourneParkvilleVic.Australia
| | - Steffen U Eisenhardt
- Department of Plastic and Hand Surgery, University of Freiburg Medical CentreMedical Faculty of the University of FreiburgFreiburgGermany
| |
Collapse
|
10
|
Dhami SPS, Patmore S, Comerford C, Byrne C, Cavanagh B, Castle J, Kirwan CC, Kenny M, Schoen I, O'Donnell JS, O'Sullivan JM. Breast cancer cells mediate endothelial cell activation, promoting von Willebrand factor release, tumor adhesion, and transendothelial migration. J Thromb Haemost 2022; 20:2350-2365. [PMID: 35722954 PMCID: PMC9796425 DOI: 10.1111/jth.15794] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/23/2022] [Accepted: 06/11/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Breast cancer results in a three- to four-fold increased risk of venous thromboembolism (VTE), which is associated with reduced patient survival. Despite this, the mechanisms underpinning breast cancer-associated thrombosis remain poorly defined. Tumor cells can trigger endothelial cell (EC) activation resulting in increased von Willebrand factor (VWF) secretion. Importantly, elevated plasma VWF levels constitute an independent biomarker for VTE risk. Moreover, in a model of melanoma, treatment with low molecular weight heparin (LMWH) negatively regulated VWF secretion and attenuated tumor metastasis. OBJECTIVE To investigate the role of VWF in breast cancer metastasis and examine the effect of LMWH in modulating EC activation and breast tumor transmigration. METHODS von Willebrand factor levels were measured by ELISA. Primary ECs were used to assess tumor-induced activation, angiogenesis, tumor adhesion, and transendothelial migration. RESULTS AND CONCLUSION Patients with metastatic breast cancer have markedly elevated plasma VWF:Ag levels that also correlate with poorer survival. MDA-MB-231 and MCF-7 breast cancer cells induce secretion of VWF, angiopoietin-2, and osteoprotegerin from ECs, which is further enhanced by the presence of platelets. Vascular endothelial growth factor-A (VEGF-A) plays an important role in modulating breast cancer-induced VWF release. Moreover, VEGF-A from breast tumor cells also contributes to a pro-angiogenic effect on ECs. VWF multimers secreted from ECs, in response to tumor-VEGF-A, mediate adhesion of breast tumor cells along the endothelium. LMWH inhibits VWF-breast tumor adhesion and transendothelial migration. Our findings highlight the significant crosstalk between tumor cells and the endothelium including increased VWF secretion which may contribute to tumor metastasis.
Collapse
Affiliation(s)
- Sukhraj Pal Singh Dhami
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| | - Sean Patmore
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| | - Claire Comerford
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| | - Ciara M. Byrne
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging CoreRoyal College of Surgeons in IrelandDublinIreland
| | - John Castle
- Manchester Cancer Research CentreThe University of ManchesterManchesterUK
| | - Cliona C. Kirwan
- Manchester Cancer Research CentreThe University of ManchesterManchesterUK
- The Nightingale CentreManchester University Foundation TrustManchester, WythenshaweUK
| | - Martin Kenny
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| | - Ingmar Schoen
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| | - James S. O'Donnell
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
- National Coagulation CentreSt James HospitalDublinIreland
| | - Jamie M. O'Sullivan
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular BiologyRoyal College of Surgeons in IrelandDublinIreland
| |
Collapse
|
11
|
Kadoglou NPE, Kapetanios D, Korakas E, Valsami G, Tentolouris N, Papanas N, Lambadiari V, Karkos C. Association of serum levels of osteopontin and osteoprotegerin with adverse outcomes after endovascular revascularisation in peripheral artery disease. Cardiovasc Diabetol 2022; 21:171. [PMID: 36050687 PMCID: PMC9438128 DOI: 10.1186/s12933-022-01605-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/30/2022] [Indexed: 12/04/2022] Open
Abstract
Background Osteoprotegerin (OPG) and osteopontin (OPN) are vascular calcification inhibitors with a known role in the atherosclerotic and inflammatory process. We investigated their relationship with adverse outcomes (restenosis/adverse cardiovascular events) after endovascular revascularisation of patients with peripheral arterial disease (PAD). Methods 203 consecutive patients were enrolled in the PAD group (PADG) and 78 age and sex-matched subjects with less than two cardiovascular risk factors served as control group (COG). PADG underwent standard medical assessment at baseline and 12 months after the procedure. During follow up major adverse cardiovascular events (MACEs) including arterial restenosis with need for reintervention were documented and the PADG was divided accordingly into two subgroups. Results During 12-month follow-up, 82 MACE were recorded (MACE subgroup). The rest of 124 PAD patients remained free of MACE (non-MACE subgroup). At baseline, OPG (9.89 ± 2.85 ng/ml vs 3.47 ± 1.95 ng/ml, p < 0.001) and OPN (79.99 ± 38.29 ng/ml vs 35.21 ± 14.84 ng/ml, p < 0.001) levels were significantly higher in PADG compared to COG, as well as in MACE subgroup compared to non-MACE subgroup (13.29 ± 3.23 ng/ml vs 10.86 ± 3 ng/ml and 96.45 ± 40.12 ng/ml vs 78.1 ± 38.29 ng/ml, respectively). An independent association of PAD with OPG and OPN was found in the whole patient cohort. Although OPG and OPN were significantly related to MACE incidence in the univariate analysis, multiple logistic regression analysis failed to detect any independent predictor of MACE within the PADG. Conclusion Baseline high OPG and OPN levels were independently associated with PAD presence. Even higher levels of those biomarkers were detected among PAD patients with MACE, however, their prognostic role should be further clarified.
Collapse
Affiliation(s)
- Nikolaos P E Kadoglou
- Medical School, University of Cyprus, 215/6 Old road Lefkosias-Lemesou, 2029, Aglatzia, Nicosia, Cyprus. .,Department of Vascular Surgery, University Hospital, LMU Munich, Munich, Germany.
| | - Dimitrios Kapetanios
- Department of Vascular Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Emmanouil Korakas
- 2nd Department of Internal Medicine, Research Institute and Diabetes Centre, Athens University Medical School, Attikon University General Hospital, Athens, Greece
| | - Georgia Valsami
- School of Health Sciences, Department of Pharmacy, National & Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Nikolaos Papanas
- Diabetes Centre, Second Department of Internal Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece
| | - Vaia Lambadiari
- 2nd Department of Internal Medicine, Research Institute and Diabetes Centre, Athens University Medical School, Attikon University General Hospital, Athens, Greece
| | - Christos Karkos
- 5Th Department of Surgery, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
12
|
Zhang X, Xu Y, Li F, Chen M. Associations between bone mineral density and subclinical peripheral arterial disease in elderly men with type 2 diabetes mellitus. Osteoporos Int 2022; 33:1715-1724. [PMID: 35451624 DOI: 10.1007/s00198-022-06404-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 04/12/2022] [Indexed: 12/24/2022]
Abstract
UNLABELLED The association between peripheral arterial disease (PAD) and osteoporosis in elderly men with type 2 diabetes mellitus (T2DM) remains unclear. We demonstrated the association between subclinical PAD and decreased total hip bone mineral density (BMD) in men aged ≥ 55 years with T2DM, providing clinical clues for the early detection of decreased bone density in total hip. PURPOSE To investigate the association between subclinical peripheral arterial disease (PAD) and bone mineral density (BMD) at total hip, femoral neck, and lumbar spine (L1-4) in elderly men with type 2 diabetes mellitus (T2DM). METHODS We identified 2,466 patients with confirmed diabetes in this retrospective cross-sectional study. A total of 272 men aged ≥ 55 years with T2DM (50 with subclinical PAD and 222 without PAD) were analyzed. Partial correlation analysis was conducted to explore the associations between ankle-brachial index (ABI) and BMD. Multivariate logistic regression analysis was performed to analyze the contributor for low bone density (T-score < - 1.0). RESULTS Patients with T2DM and subclinical PAD (ABI ≤ 0.9) had significantly lower total hip BMD and T-score (BMD, 0.87 ± 0.14 vs. 0.92 ± 0.15 g/cm2, P = 0.014; T-score, -1.30 [-1.70 to -0.45] vs. -0.80 [-1.40 to 0.00], P = 0.009) than those in the control group. The partial correlation analyses indicated that ABI significantly correlated with the total hip T-score (adjusted r = 0.166, P = 0.009). The logistic regression analysis indicated that subclinical PAD was an independent risk factor for the risk of decreased bone density in total hip (adjusted odds ratio [95% CI]: 8.933 [1.075-74.222], P = 0.043). CONCLUSION: Subclinical PAD (ABI ≤ 0.9) could be used as a risk factor for decreased total hip BMD in men aged ≥ 55 years with T2DM, which provides clinical clues for the early detection of low bone density in total hip in such populations.
Collapse
Affiliation(s)
- X Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Y Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - F Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - M Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
13
|
Peyronnel C, Totoson P, Petitcolin V, Bonnefoy F, Guillot X, Saas P, Verhoeven F, Martin H, Demougeot C. Effects of local cryotherapy on systemic endothelial activation, dysfunction, and vascular inflammation in adjuvant-induced arthritis (AIA) rats. Arthritis Res Ther 2022; 24:97. [PMID: 35488311 PMCID: PMC9052534 DOI: 10.1186/s13075-022-02774-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
Aim This study explored the systemic vascular effects of local cryotherapy with a focus on endothelial changes and arterial inflammation in the model of rat adjuvant-induced arthritis (AIA). Methods Cryotherapy was applied twice a day on hind paws of AIA rats from the onset of arthritis to the acute inflammatory phase. Endothelial activation was studied in the aorta by measuring the mRNA levels of chemokines (CXCL-1, MCP-1 (CCL-2), MIP-1α (CCL-3)) and adhesion molecules (ICAM-1, VCAM-1) by qRT-PCR. Endothelial dysfunction was measured in isolated aortic and mesenteric rings. Aortic inflammation was evaluated via the mRNA expression of pro-inflammatory cytokines (TNF-α, IL-6) by qRT-PCR and leucocyte infiltration analysis (flow cytometry). Plasma levels of TNF-α, IL-6, IL-1β, IL-17A, and osteoprotegerin (OPG) were measured using Multiplex/ELISA. Results AIA was associated with an increased aortic expression of CXCL-1 and ICAM-1 as well as an infiltration of leucocytes and increased mRNA expression of IL-6, IL-1β, and TNF-α. Local cryotherapy, which decreased arthritis score and structural damages, reduced aortic mRNA expression of CXCL-1, IL-6, IL-1β, and TNF-α, as well as aortic infiltration of leucocytes (T lymphocytes, monocytes/macrophages, neutrophils) and improved acetylcholine-induced vasorelaxation in the aorta and mesenteric arteries. Plasma levels of IL-17A and OPG were significantly reduced by cryotherapy, while the number of circulating leucocytes was not. IL-17A levels positively correlated with endothelial activation and dysfunction. Conclusion In the AIA model, local cryotherapy reduced systemic endothelial activation, immune cell infiltration, and endothelial dysfunction. Mechanistically, the reduction of circulating levels of IL-17A appears as the possible link between joint cooling and the remote vascular effects. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02774-1.
Collapse
Affiliation(s)
- C Peyronnel
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - P Totoson
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - V Petitcolin
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - F Bonnefoy
- INSERM UMR 1098 RIGHT, EFS BFC, Univ. Bourgogne Franche-Comté, LabEX LipSTIC, F-25000, Besançon, France
| | - X Guillot
- Service de Rhumatologie, CHU Felix Guyon, Ile de la Réunion, Saint-Denis, France
| | - P Saas
- INSERM UMR 1098 RIGHT, EFS BFC, Univ. Bourgogne Franche-Comté, LabEX LipSTIC, F-25000, Besançon, France
| | - F Verhoeven
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France.,Service de Rhumatologie, CHRU Besançon, F-25000, Besançon, France
| | - H Martin
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - C Demougeot
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France.
| |
Collapse
|
14
|
Williams PT. Quantile-Specific Heritability of Inflammatory and Oxidative Stress Biomarkers Linked to Cardiovascular Disease. J Inflamm Res 2022; 15:85-103. [PMID: 35023945 PMCID: PMC8743501 DOI: 10.2147/jir.s347402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Heritability (h2 , the proportion of the phenotypic variance attributable to additive genetic effects) is traditionally assumed to be constant throughout the distribution of the phenotype. However, the heritabilities of circulating C-reactive protein, interleukin-6, plasminogen activator inhibitor type-1 (PAI-1), and monocyte chemoattractant protein-1 (MCP-1) concentrations depend upon whether the phenotype is high or low relative to their distributions (quantile-dependent expressivity), which may account for apparent gene-environment interactions. Whether the heritabilities of other inflammatory biomarkers linked to cardiovascular disease are quantile-dependent remain to be determined. PATIENTS AND METHODS Quantile-specific offspring-parent (βOP) and full-sib regression slopes (βFS) were estimated by applying quantile regression to the age- and sex-adjusted phenotypes of families surveyed as part of the Framingham Heart Study. Quantile-specific heritabilities were calculated as: h2 =2βOP/(1+rspouse) and h2 ={(1+8rspouseβFS)0.5-1}/(2rspouse). RESULTS Heritability (h2 ± SE) of lipoprotein-associated phospholipase A2 (Lp-PLA2) mass concentrations increased from 0.11 ± 0.03 at the 10th percentile, 0.08 ± 0.03 at the 25th, 0.12 ± 0.03 at the 50th, 0.20 ± 0.04 at the 75th, and 0.26 ± 0.06 at the 90th percentile, or 0.0023 ± 0.0006 per each one-percent increase in the phenotype distribution (Plinear trend= 0.0004). Similarly, h2 increased 0.0029 ± 0.0011 (Plinear trend= 0.01) for sP-selectin, 0.0032 ± 0.0009 (Plinear trend= 0.0001) for soluble intercellular adhesion molecule 1 (sICAM-1), and 0.0026 ± 0.0006 for tumor necrosis factor receptor 2 (TNFR2) (Plinear trend= 5.0 × 10-6) per each one-percent increase in their distributions when estimated from βOP. Osteoprotegerin and soluble ST2 heritability also increased significantly with increasing percentiles of their distributions when estimated from βFS. Lp-PLA2 activity, CD40 ligand, TNFα, interleukin-18, and myeloperoxidase heritability showed no significant quantile-dependence. CONCLUSION The heritabilities of circulating Lp-PLA2-mass, sP-selectin, sICAM-1, TNFR2, osteoprotegerin and soluble ST2 concentrations are quantile-dependent, which may contribute to purported genetic modulations of: 1) sP-selectin's relationships to venous thrombosis, pulmonary hypertension, type 2 diabetes and atorvastatin treatment; 2) sICAM-I's relationships to brain abscess and atorvastatin treatment; and 3) Lp-PLA2's relationships to myocardial infarction and preeclampsia.
Collapse
Affiliation(s)
- Paul T Williams
- Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
15
|
Celebi G, Anapali M, Dagistanli FK, Akdemir AS, Aydemir D, Ulusu NN, Ulutin T, Komurcu-Bayrak E. Effect of vitamin D supplementation on OPG/RANKL signalling activities in endothelial tissue damage in diet-induced diabetic rat model. Pharmacol Rep 2021; 74:124-134. [PMID: 34657267 DOI: 10.1007/s43440-021-00332-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Type 2 Diabetes Mellitus is a chronic metabolic disease that causes endothelial damage and is an important risk factor for atherosclerosis. In the present study vitamin D3 supplementation in rats was used to determine the role of Osteoprotegerin (OPG)/Receptor activator kB ligand (RANKL) signalling in endothelial damage and changes in the expression levels of genes involved in this pathway. We hypothesized that vitamin D3 supplementation affects OPG and RANKL activity in the aorta. METHODS Diabetes was induced in rats via injections of 40 mg/kg of streptozotocin followed by a high fructose (10%) diet. Group 2 (healthy) and 4 (diabetic) received 170 IU/kg of vitamin D3 weekly for 5 weeks, while Group 1 (healthy) and 2 (diabetic) received sterile saline. The aortas of each group were collected to analyse mRNA expression using the real-time PCR method and also to evaluate magnesium and calcium levels using inductively coupled plasma mass spectrometry. RESULTS Opg and Il-1b expression levels were significantly associated with both diabetes and vitamin D3 supplementation in the aortas of the study groups (p ≤ 0.05). Opg mRNA expression was also found to correlate with both Icam-1 and Nos3 mRNA expression levels (r = 0.699, p = 0.001 and r = 0.622, p = 0.003, respectively). In addition, when mineral levels in the aortic tissues were compared among all groups, it was found that the interaction of diabetes and vitamin D3 supplementation significantly affected Mg levels and Mg/Ca ratios. CONCLUSIONS It is concluded that vitamin D3 supplementation has a modulatory effect on OPG/RANKL activity in the vessel wall by ameliorating endothelial damage in diabetes. This effect may contribute to the regulation of cytokine-mediated vascular homeostasis and mineral deposition in the aorta; therefore, further comprehensive studies are proposed to demonstrate this relationship.
Collapse
Affiliation(s)
- Gizem Celebi
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey. .,Graduate School of Health Sciences, Istanbul University, Istanbul, Turkey. .,Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, 34956, Istanbul, Turkey.
| | - Merve Anapali
- Cerrahpasa Medical Faculty, Medical Biology Department, Istanbul University-Cerrahpasa, Istanbul, Turkey.,Medical Biology Department, Ataturk University Medical Faculty, Erzurum, Turkey
| | - Fatma Kaya Dagistanli
- Cerrahpasa Medical Faculty, Medical Biology Department, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ayse Seda Akdemir
- Cerrahpasa Medical Faculty, Medical Biology Department, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Duygu Aydemir
- School of Medicine, Department of Medical Biochemistry, Koç University, 34450, Sariyer, Istanbul, Turkey.,Koç University Research Center for Translational Medicine (KUTTAM), 34450, Sariyer, Istanbul, Turkey
| | - Nuriye Nuray Ulusu
- School of Medicine, Department of Medical Biochemistry, Koç University, 34450, Sariyer, Istanbul, Turkey.,Koç University Research Center for Translational Medicine (KUTTAM), 34450, Sariyer, Istanbul, Turkey
| | - Turgut Ulutin
- Cerrahpasa Medical Faculty, Medical Biology Department, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Evrim Komurcu-Bayrak
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.,Istanbul Faculty of Medicine, Department of Medical Genetics, Istanbul University, Istanbul, Turkey
| |
Collapse
|
16
|
Dutka M, Bobiński R, Wojakowski W, Francuz T, Pająk C, Zimmer K. Osteoprotegerin and RANKL-RANK-OPG-TRAIL signalling axis in heart failure and other cardiovascular diseases. Heart Fail Rev 2021; 27:1395-1411. [PMID: 34313900 PMCID: PMC9197867 DOI: 10.1007/s10741-021-10153-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2021] [Indexed: 01/29/2023]
Abstract
Osteoprotegerin (OPG) is a glycoprotein involved in the regulation of bone remodelling. OPG regulates osteoclast activity by blocking the interaction between the receptor activator of nuclear factor kappa B (RANK) and its ligand (RANKL). More and more studies confirm the relationship between OPG and cardiovascular diseases. Numerous studies have confirmed that a high plasma concentration of OPG and a low concentration of tumour necrosis factor–related apoptosis inducing ligand (TRAIL) together with a high OPG/TRAIL ratio are predictors of poor prognosis in patients with myocardial infarction. A high plasma OPG concentration and a high ratio of OPG/TRAIL in the acute myocardial infarction are a prognostic indicator of adverse left ventricular remodelling and of the development of heart failure. Ever more data indicates the participation of OPG in the regulation of the function of vascular endothelial cells and the initiation of the atherosclerotic process in the arteries. Additionally, it has been shown that TRAIL has a protective effect on blood vessels and exerts an anti-atherosclerotic effect. The mechanisms of action of both OPG and TRAIL within the cells of the vascular wall are complex and remain largely unclear. However, these mechanisms of action as well as their interaction in the local vascular environment are of great interest to researchers. This article presents the current state of knowledge on the mechanisms of action of OPG and TRAIL in the circulatory system and their role in cardiovascular diseases. Understanding these mechanisms may allow their use as a therapeutic target in cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland.
| | - Rafał Bobiński
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Wojciech Wojakowski
- Department of Cardiology and Structural Heart Disease, Medical University of Silesia, Katowice, Poland
| | - Tomasz Francuz
- Department of Biochemistry, Medical University of Silesia, Katowice, Poland
| | - Celina Pająk
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Karolina Zimmer
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, University of Bielsko-Biala, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| |
Collapse
|
17
|
The role of osteoprotegerin (OPG) in fibrosis: its potential as a biomarker and/or biological target for the treatment of fibrotic diseases. Pharmacol Ther 2021; 228:107941. [PMID: 34171336 DOI: 10.1016/j.pharmthera.2021.107941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Fibrosis is defined by excessive formation and accumulation of extracellular matrix proteins, produced by myofibroblasts, that supersedes normal wound healing responses to injury and results in progressive architectural remodelling. Fibrosis is often detected in advanced disease stages when an organ is already severely damaged and can no longer function properly. Therefore, there is an urgent need for reliable and easily detectable markers to identify and monitor fibrosis onset and progression as early as possible; this will greatly facilitate the development of novel therapeutic strategies. Osteoprotegerin (OPG), a well-known regulator of bone extracellular matrix and most studied for its role in regulating bone mass, is expressed in various organs and functions as a decoy for receptor activator of nuclear factor kappa-B ligand (RANKL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Recently, OPG has been linked to fibrosis and fibrogenesis, and has been included in a panel of markers to diagnose liver fibrosis. Multiple studies now suggest that OPG may be a general biomarker suitable for detection of fibrosis and/or monitoring the impact of fibrosis treatment. This review summarizes our current understanding of the role of OPG in fibrosis and will discuss its potential as a biomarker and/or novel therapeutic target for fibrosis.
Collapse
|
18
|
Deligiorgi MV, Panayiotidis MI, Siasos G, Trafalis DT. Osteoporosis Entwined with Cardiovascular Disease: The Implication of Osteoprotegerin and the Example of Statins. Curr Med Chem 2021; 28:1443-1467. [PMID: 31971101 DOI: 10.2174/0929867327666200123151132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 11/22/2022]
Abstract
Beyond being epiphenomenon of shared epidemiological factors, the integration of Osteoporosis (OP) with Cardiovascular Disease (CVD) - termed "calcification paradox" - reflects a continuum of aberrant cardiometabolic status. The present review provides background knowledge on "calcification paradox", focusing on the endocrine aspect of vasculature orchestrated by the osteoblastic molecular fingerprint of vascular cells, acquired via imbalance among established modulators of mineralization. Osteoprotegerin (OPG), the well-established osteoprotective cytokine, has recently been shown to exert a vessel-modifying role. Prompted by this notion, the present review interrogates OPG as the potential missing link between OP and CVD. However, so far, the confirmation of this hypothesis is hindered by the equivocal role of OPG in CVD, being both proatherosclerotic and antiatherosclerotic. Further research is needed to illuminate whether OPG could be a biomarker of the "calcification paradox". Moreover, the present review brings into prominence the dual role of statins - cardioprotective and osteoprotective - as a potential illustration of the integration of CVD with OP. Considering that the statins-induced modulation of OPG is central to the statins-driven osteoprotective signalling, statins could be suggested as an illustration of the role of OPG in the bone/vessels crosstalk, if further studies consolidate the contribution of OPG to the cardioprotective role of statins. Another outstanding issue that merits further evaluation is the inconsistency of the osteoprotective role of statins. Further understanding of the varying bone-modifying role of statins, likely attributed to the unique profile of different classes of statins defined by distinct physicochemical characteristics, may yield tangible benefits for treating simultaneously OP and CVD.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology - Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens, Building 16, 1st Floor, 75 Mikras Asias, 11527 Goudi, Athens, Greece
| | - Mihalis I Panayiotidis
- Department of Applied Sciences, Group of Translational Biosciences, Faculty of Health & Life Sciences, Northumbria University, Ellison Building A516, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| | - Gerasimos Siasos
- Department of Cardiology, Faculty of Medicine, 1st Hippokration Hospital, National and Kapodistrian University of Athens, 114 Vas Sofias, 11527 Athens, Greece
| | - Dimitrios T Trafalis
- Department of Pharmacology - Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens, Building 16, 1st Floor, 75 Mikras Asias, 11527 Goudi, Athens, Greece
| |
Collapse
|
19
|
Zhang Q, Chen T, Zhang Y, Lyu L, Zhang B, Huang C, Zhou X, Wu Y, Li Z. MiR-30c-5p regulates adventitial progenitor cells differentiation to vascular smooth muscle cells through targeting OPG. Stem Cell Res Ther 2021; 12:67. [PMID: 33468212 PMCID: PMC7814722 DOI: 10.1186/s13287-020-02127-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/28/2020] [Indexed: 11/28/2022] Open
Abstract
Background As the most important component of the vascular wall, vascular smooth muscle cells (VSMCs) participate in the pathological process by phenotype transformation or differentiation from stem/progenitor cells. The main purpose of this study was to reveal the role and related molecular mechanism of microRNA-30c-5p (miR-30c-5p) in VSMC differentiation from adventitial progenitor cells expressing stem cell antigen-1(Sca-1). Methods In this study, we detected the expression of miR-30c-5p in human normal peripheral arteries and atherosclerotic arteries. In vitro, a stable differentiation model from adventitial Sca-1+ progenitor cells to VSMCs was established using transforming growth factor-β1 (TGF-β1) induction and the expression of miR-30c-5p during the process was observed. Then, we explored the effect of miR-30c-5p overexpression and inhibition on the differentiation from adventitial Sca-1+ progenitor cells to VSMCs. The target genes of miR-30c-5p were identified by protein chip and biological analyses and the expression of these genes in the differentiation process were detected. Further, the relationship between the target gene and miR-30c-5p and its effect on differentiation were evaluated. Finally, the co-transfection of miR-30c-5p inhibitor and small interfering RNA (siRNA) of the target gene was implemented to verify the functional target gene of miR-30c-5p during the differentiation from adventitial Sca-1+ progenitor cells to VSMCs, and the dual-luciferase reporter gene assay was performed to detect whether the mRNA 3′untranslated region (UTR) of the target gene is the direct binding site of miR-30c-5p. Results The expression of miR-30c-5p in the human atherosclerotic arteries was significantly lower than that in the normal arteries. During the differentiation from adventitial Sca-1+ progenitor cells to VSMCs, the expression of VSMC special markers including smooth muscle α-actin (SMαA), smooth muscle-22α (SM22α), smooth muscle myosin heavy chain (SMMHC), and h1-caponin increased accompanied with cell morphology changing from elliptic to fusiform. Meanwhile, the expression of miR-30c-5p decreased significantly. In functional experiments, overexpression of miR-30c-5p inhibited SMαA, SM22α, SMMHC, and h1-caponin at the mRNA and protein levels. In contrast, inhibition of miR-30c-5p promoted the expression of SMαA, SM22α, SMMHC, and h1-caponin. The target gene, osteoprotegerin (OPG), was predicted through protein chip and bioinformatics analyses. Overexpression of miR-30c-5p inhibited OPG expression while inhibition of miR-30c-5p had an opposite effect. Co-transfection experiments showed that low expression of OPG could weaken the promotion effect of miR-30c-5p inhibitor on the differentiation from adventitial Sca-1+ progenitor cells to VSMCs and the dual-luciferase reporter gene assay demonstrated that miR-30c-5p could target the mRNA 3′UTR of OPG directly. Conclusions This study demonstrates that miR-30c-5p expression was significantly decreased in atherosclerotic arteries and miR-30c-5p inhibited VSMC differentiation from adventitial Sca-1+ progenitor cells through targeting OPG, which may provide a new target for the treatment of VSMCs-associated diseases.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Yun Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Lingxia Lyu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Bohuan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Chengchen Huang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Xuhao Zhou
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Yutao Wu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China.
| | - Zhoubin Li
- Department of Lung Transplantation, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China.
| |
Collapse
|
20
|
Cetin Z, Berker D, Okudan B, Kilinckaya M, Turhan T, Catak M. The role of serum osteoprotegerin level in diagnosis of disease and determining cardiovascular risk of polycystic ovary syndrome. Gynecol Endocrinol 2020; 36:943-946. [PMID: 32338102 DOI: 10.1080/09513590.2020.1754786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Objective: Although animal studies claim that osteoprotegerin (OPG) is preventive on this system, there are conflicting results in human studies. The aim of this study was to investigate the role of OPG in the diagnosis and determination of cardivovascular risk in patients with polycystic ovary syndrome (PCOS), which is a multisystem effective disease.Method: The study was performed on 28 premenopausal healthy female volunteers and 57 newly diagnosed PCOS patients in 2017. Anamnesis was obtained, body mass indexes were calculated, laboratory tests required for diagnosis and differential diagnosis of PCOS and suprapubic ovarian ultrasonography were performed, serum OPG level was studied by enzyme-linked immunosorbent assay.Results: OPG levels were similar in PCOS and control groups and there was no significant difference (49.392 ± 10.973 pg/ml vs 49.567 ± 13.57 pg/ml, p = .815). Correlation analysis showed a positive correlation between OPG and total testosterone levels in the PCOS group (r = 0.277, p = .045). No significant relationship with cardiovascular and metabolic parameters was detected.Conclusion: No difference was found between PCOS patients and control groups in terms of OPG levels. Therefore, it is thought that OPG level cannot be used in the diagnosis of the disease. There was no significant relationship between cardiometabolic parameters.
Collapse
Affiliation(s)
- Zeynep Cetin
- Endocrinology and Metabolism Department, Amasya Üniversity Sabuncuoğlu Şerefeddin Education and Research Hospital, Amasya, Turkey
| | - Dilek Berker
- Endocrinology and Metabolism Department, Ankara City Hospital, Ankara, Turkey
| | - Berna Okudan
- Nuclear Medicine Department, Ankara City Hospital, Ankara, Turkey
| | | | - Turan Turhan
- Biochemistry Department, Ankara City Hospital, Ankara, Turkey
| | - Merve Catak
- Endocrinology and Metabolism Department, Tokat Public Hospital, Tokat, Turkey
| |
Collapse
|
21
|
Early Endothelial Activation Precedes Glycocalyx Degradation and Microvascular Dysfunction in Experimentally Induced Plasmodium falciparum and Plasmodium vivax Infection. Infect Immun 2020; 88:IAI.00895-19. [PMID: 32122938 DOI: 10.1128/iai.00895-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
Endothelial activation and microvascular dysfunction are key pathogenic processes in severe malaria. We evaluated the early role of these processes in experimentally induced Plasmodium falciparum and P. vivax infection. Participants were enrolled in induced blood-stage malaria clinical trials. Plasma osteoprotegerin, angiopoietin-2, and von Willebrand Factor (vWF) levels were measured as biomarkers of endothelial activation. Microvascular function was assessed using peripheral arterial tonometry and near-infrared spectroscopy, and the endothelial glycocalyx was assessed by sublingual videomicroscopy and measurement of biomarkers of degradation. Forty-five healthy, malaria-naive participants were recruited from 5 studies. Osteoprotegerin and vWF levels increased in participants following inoculation with P. vivax (n = 16) or P. falciparum (n = 15), with the angiopoietin-2 level also increasing in participants following inoculation with P. falciparum For both species, the most pronounced increase was seen in osteoprotegerin. This was particularly marked in participants inoculated with P. vivax, where the osteoprotegerin level correlated with the levels of parasitemia and the malaria clinical score. There were no changes in measures of endothelial glycocalyx or microvascular function. Plasma biomarkers of endothelial activation increased in early P. falciparum and P. vivax infection and preceded changes in the endothelial glycocalyx or microvascular function. The more pronounced increase in osteoprotegerin suggests that this biomarker may play a role in disease pathogenesis.
Collapse
|
22
|
Plasma Osteoprotegerin Correlates with Stroke Severity and the Occurrence of Microembolic Signals in Patients with Acute Ischemic Stroke. DISEASE MARKERS 2019; 2019:3090364. [PMID: 31191747 PMCID: PMC6525837 DOI: 10.1155/2019/3090364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 03/07/2019] [Accepted: 04/01/2019] [Indexed: 12/20/2022]
Abstract
Background Instability of atherosclerotic plaques is associated with the occurrence of stroke. Microembolic signals (MESs) are an indicator of unstable plaque. A relationship between plasma osteoprotegerin (OPG) and ischemic stroke has already been identified. The aim of this study was to investigate whether plasma OPG levels have a relationship with MESs and to evaluate the feasibility of OPG as a biomarker of stroke severity and occurrence of MESs. Methods Our study consisted of 127 patients with large artery atherosclerosis stroke and 56 controls. Patients were classified into subgroups based on stroke severity and the occurrence of MESs. MES-monitoring was performed for 60 min using transcranial Doppler within 72 h of stroke onset. Stroke severity at admission was assessed by the National Institutes of Health Stroke Scale. Results Plasma OPG levels were significantly associated with stroke, MESs, and stroke severity at admission (adjusted OR [95% CI]: 1.002 [1.001–1.003] p < 0.001; 1.002 [1.001–1.003] p = 0.001; 1.001 [1.000–1.002] p = 0.028). When plasma OPG levels were used to determine the stroke severity, the area under the receiver-operating characteristic curve (AUC) was 0.734 (95% CI: 0.625-0.843) based on a cutoff value of 1998.44 pg/ml; the sensitivity and specificity of this test were 80.6% and 65.6%, respectively. Furthermore, when the levels of OPG were used to distinguish the presence of MESs, the AUC was 0.766 (95% CI: 0.672-0.860); the cutoff value was 2107.91 pg/ml. The sensitivity of this cutoff value was 68.8% and the specificity was 73.7%. Conclusions Plasma OPG levels correlate with stroke severity and the occurrence of MESs.
Collapse
|
23
|
The Role of Osteoprotegerin and Its Ligands in Vascular Function. Int J Mol Sci 2019; 20:ijms20030705. [PMID: 30736365 PMCID: PMC6387017 DOI: 10.3390/ijms20030705] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/01/2019] [Accepted: 02/02/2019] [Indexed: 12/15/2022] Open
Abstract
The superfamily of tumor necrosis factor (TNF) receptors includes osteoprotegerin (OPG) and its ligands, which are receptor activators of nuclear factor kappa-B ligand (RANKL) and TNF-related apoptosis-inducing ligand (TRAIL). The OPG/RANKL/RANK system plays an active role in pathological angiogenesis and inflammation as well as cell survival. It has been demonstrated that there is crosstalk between endothelial cells and osteoblasts during osteogenesis, thus establishing a connection between angiogenesis and osteogenesis. This OPG/RANKL/RANK/TRAIL system acts on specific cell surface receptors, which are then able to transmit their signals to other intracellular components and modify gene expression. Cytokine production and activation of their receptors induce mechanisms to recruit monocytes and neutrophils as well as endothelial cells. Data support the role of an increased OPG/RANKL ratio as a possible marker of progression of endothelial dysfunction in metabolic disorders in relationship with inflammatory marker levels. We review the role of the OPG/RANKL/RANK triad in vascular function as well as molecular mechanisms related to the etiology of vascular diseases. The potential therapeutic strategies may be very promising in the future.
Collapse
|
24
|
Tschiderer L, Klingenschmid G, Nagrani R, Willeit J, Laukkanen JA, Schett G, Kiechl S, Willeit P. Osteoprotegerin and Cardiovascular Events in High-Risk Populations: Meta-Analysis of 19 Prospective Studies Involving 27 450 Participants. J Am Heart Assoc 2018; 7:e009012. [PMID: 30369329 PMCID: PMC6201389 DOI: 10.1161/jaha.118.009012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/16/2018] [Indexed: 01/17/2023]
Abstract
Background Osteoprotegerin is a cytokine involved in bone metabolism as well as vascular calcification and atherogenesis. Although circulating osteoprotegerin levels are robustly associated with incident cardiovascular disease ( CVD ) in the general population, its relevance as a biomarker among populations at high CVD risk is less clear. Methods and Results Three independent reviewers systematically searched PubMed, EMBASE , and Web of Science to identify prospective studies that had recruited participants on the basis of having conditions related to high CVD risk. A total of 19 studies were eligible for inclusion, reporting on 27 450 patients with diabetes mellitus (2 studies), kidney disease (7 studies), preexisting heart disease (5 studies), or recent acute coronary syndromes (5 studies) at baseline. Over a mean follow-up of 4.2 years, 4066 CVD events were recorded. In a random-effects meta-analysis, the pooled risk ratio for CVD events comparing people in the top versus the bottom tertile of osteoprotegerin concentration was 1.30 (95% confidence interval, 1.12-1.50; P<0.001; I2=68.3%). There was evidence for presence of publication bias ( P value from Egger's test=0.013). Correction for publication bias using the trim-and-fill method reduced the risk ratio to 1.21 (95% confidence interval, 1.03-1.42; P<0.001). The risk ratios did not vary significantly by population type, geographical region, statistical adjustment, sample or assay type, age, sex, or length of follow-up. Conclusions In populations at high CVD risk, elevated circulating osteoprotegerin levels are associated with a higher risk for future CVD events. The magnitude of association appears weaker than in the general population.
Collapse
Affiliation(s)
- Lena Tschiderer
- Department of NeurologyMedical University of InnsbruckAustria
| | | | - Rajini Nagrani
- Department of NeurologyMedical University of InnsbruckAustria
| | - Johann Willeit
- Department of NeurologyMedical University of InnsbruckAustria
| | - Jari A. Laukkanen
- Institute of Public Health and Clinical NutritionUniversity of Eastern FinlandKuopioFinland
- Central Finland Central HospitalJyväskyläFinland
- Faculty of Sport and Health SciencesUniversity of JyväskyläFinland
| | - Georg Schett
- Department of Internal Medicine 3University of Erlangen‐NurembergErlangenGermany
| | - Stefan Kiechl
- Department of NeurologyMedical University of InnsbruckAustria
| | - Peter Willeit
- Department of NeurologyMedical University of InnsbruckAustria
- Department of Public Health and Primary CareUniversity of CambridgeUnited Kingdom
| |
Collapse
|
25
|
Kamimura D, Suzuki T, Furniss AL, Griswold ME, Kullo IJ, Lindsey ML, Winniford MD, Butler KR, Mosley TH, Hall ME. Elevated serum osteoprotegerin is associated with increased left ventricular mass index and myocardial stiffness. J Cardiovasc Med (Hagerstown) 2018; 18:954-961. [PMID: 28787318 DOI: 10.2459/jcm.0000000000000549] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
AIM Osteoprotegerin (OPG) is associated with a poor prognosis in patients with heart failure with preserved ejection fraction (HFpEF). OPG has also been associated with fibrosis and collagen cross-linking, which increase arterial and left ventricle (LV) myocardial stiffness. Little is known about the relation of OPG and LV structure and function in African-Americans who are disproportionately affected by HFpEF. METHODS AND RESULTS Our analysis included 1172 participants with preserved LV ejection fraction (>50%) from the African-American cohort in the Genetic Epidemiology Network of Arteriopathy Study (mean age 63 years, 72% female). We used diastolic wall strain indicator measured by echocardiography to assess LV myocardial stiffness. Diastolic wall strain was calculated as (LV posterior thickness at end-systole - LV posterior thickness at end-diastole)/LV posterior thickness at end-systole. Associations between OPG levels and indices of arterial and LV structure and function were evaluated by using generalized linear mixed models and adjusted for possible confounders. OPG levels were correlated with age, female sex, presence of hypertension and diabetes, and lower estimated glomerular filtration rate (P < 0.05 for all). Multivariable analysis revealed that higher OPG levels were associated with greater LV mass index, increased LV myocardial stiffness, and higher N-terminal prohormone brain natriuretic peptide levels (P < 0.05 for all). CONCLUSION In African-Americans, higher OPG levels were associated with characteristics common in patients with HFpEF and were significantly associated with known precursors to HFpEF. These findings indicate a potential role for OPG in the pathophysiology of HFpEF in African-Americans.
Collapse
Affiliation(s)
- Daisuke Kamimura
- aDivision of CardiologybDepartment of Medicine, Center for Biostatistics and Bioinformatics, University of Mississippi Medical Center, Jackson, MississippicDivision of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MinnesotadDepartment of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical CentereResearch Service, G.V. (Sonny) Montgomery Veterans Affairs Medical CenterfDivision of Geriatric Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Barber BE, Grigg MJ, Piera KA, William T, Cooper DJ, Plewes K, Dondorp AM, Yeo TW, Anstey NM. Intravascular haemolysis in severe Plasmodium knowlesi malaria: association with endothelial activation, microvascular dysfunction, and acute kidney injury. Emerg Microbes Infect 2018; 7:106. [PMID: 29872039 PMCID: PMC5988665 DOI: 10.1038/s41426-018-0105-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 12/29/2022]
Abstract
Plasmodium knowlesi occurs throughout Southeast Asia, and is the most common cause of human malaria in Malaysia. Severe disease in humans is characterised by high parasite biomass, reduced red blood cell deformability, endothelial activation and microvascular dysfunction. However, the roles of intravascular haemolysis and nitric oxide (NO)-dependent endothelial dysfunction, important features of severe falciparum malaria, have not been evaluated, nor their role in acute kidney injury (AKI). In hospitalised Malaysian adults with severe (n = 48) and non-severe (n = 154) knowlesi malaria, and in healthy controls (n = 50), we measured cell-free haemoglobin (CFHb) and assessed associations with the endothelial Weibel–Palade body (WPB) constituents, angiopoietin-2 and osteoprotegerin, endothelial and microvascular function, and other markers of disease severity. CFHb was increased in knowlesi malaria in proportion to disease severity, and to a greater extent than previously reported in severe falciparum malaria patients from the same study cohort. In knowlesi malaria, CFHb was associated with parasitaemia, and independently associated with angiopoietin-2 and osteoprotegerin. As with angiopoietin-2, osteoprotegerin was increased in proportion to disease severity, and independently associated with severity markers including creatinine, lactate, interleukin-6, endothelial cell adhesion molecules ICAM-1 and E-selectin, and impaired microvascular reactivity. Osteoprotegerin was also independently associated with NO-dependent endothelial dysfunction. AKI was found in 88% of those with severe knowlesi malaria. Angiopoietin-2 and osteoprotegerin were both independent risk factors for acute kidney injury. Our findings suggest that haemolysis-mediated endothelial activation and release of WPB constituents is likely a key contributor to end-organ dysfunction, including AKI, in severe knowlesi malaria.
Collapse
Affiliation(s)
- Bridget E Barber
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, NT, Australia. .,Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Sabah, Malaysia.
| | - Matthew J Grigg
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, NT, Australia.,Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Sabah, Malaysia
| | - Kim A Piera
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, NT, Australia
| | - Timothy William
- Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Sabah, Malaysia.,Jesselton Medical Centre, Kota Kinabalu, Sabah, Malaysia.,Clinical Research Centre, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
| | - Daniel J Cooper
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, NT, Australia.,Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Sabah, Malaysia
| | - Katherine Plewes
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Arjen M Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Tsin W Yeo
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, NT, Australia.,Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Sabah, Malaysia.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Institute of Infectious Disease and Epidemiology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Nicholas M Anstey
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, NT, Australia.,Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Sabah, Malaysia
| |
Collapse
|
27
|
Rochette L, Meloux A, Rigal E, Zeller M, Cottin Y, Vergely C. The role of osteoprotegerin in the crosstalk between vessels and bone: Its potential utility as a marker of cardiometabolic diseases. Pharmacol Ther 2018; 182:115-132. [DOI: 10.1016/j.pharmthera.2017.08.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
28
|
Nugroho J, Widorini W. Correlation between Osteoprotegerin Serum Level and Coronary Calcification Using Coronary Artery Calcium Score in Patient with Moderate-Severe Cardiovascular Risk Factor. Int J Angiol 2017; 26:234-237. [PMID: 29142489 DOI: 10.1055/s-0037-1607050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Coronary artery calcification is a part of atherosclerosis process associated with coronary heart disease. Recently, coronary artery calcification assessment using computed tomography (CT) is still the best noninvasive imaging with high sensitivity and specificity. Osteoprotegerin (OPG) is one of vascular calcification marker that through its role to bind receptor activator of nuclear factor-κβ ligand and inhibit osteoclastogenesis is suspected of playing a role for coronary calcification in atherosclerosis process. The objective of this study was to prove a positive correlation between OPG serum level and coronary calcification using coronary artery calcium (CAC) score in patient with moderate-severe cardiovascular (CV) risk factor. This is a cross-sectional study with purposive sampling technique. Thirty-three subjects participate in this research and each subject underwent a multislice computed tomography (MSCT) examination to assess coronary calcification and their blood samples were collected for OPG measurement. This study is analyzed with Spearman's correlation test. The mean of OPG serum level in this study was 5.89 ± 2.1 pmol/L for moderate-risk Framingham risk score (FRS) and the mean of OPG serum level for high-risk FRS was 7.27 ± 3.4. There was a positive, moderate, and significant correlation between OPG serum level and coronary calcification using CAC score in patient with moderate-severe CV risk factor ( r = 0.694; p < 0.001).
Collapse
Affiliation(s)
- J Nugroho
- Department of Cardiology and Vascular Medicine, Soetomo General Hospital, Airlangga University, Surabaya, East Java, Indonesia
| | - Widorini Widorini
- Department of Cardiology and Vascular Medicine, Soetomo General Hospital, Airlangga University, Surabaya, East Java, Indonesia
| |
Collapse
|
29
|
Abu El-Asrar AM, Ahmad A, Alam K, Bittoun E, Siddiquei MM, Mohammad G, Mousa A, De Hertogh G, Opdenakker G. Unbalanced Vitreous Levels of Osteoprotegerin, RANKL, RANK, and TRAIL in Proliferative Diabetic Retinopathy. Ocul Immunol Inflamm 2017; 26:1248-1260. [PMID: 28914577 DOI: 10.1080/09273948.2017.1343855] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE We investigated the expression of the proinflammatory and proangiogenic factor osteoprotegerin (OPG) and its ligands, receptor activator of nuclear factor-κB ligand (RANKL), tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), and the receptor RANK in proliferative diabetic retinopathy (PDR). MATERIALS AND METHODS Vitreous samples from PDR and nondiabetic control patients and epiretinal membranes from PDR patients were studied by enzyme-linked immunosorbent assay, immunohistochemistry, and Western blot analysis. RESULTS Vascular endothelial growth factor, OPG, and soluble RANK levels in vitreous samples from PDR patients were significantly higher than that in nondiabetic controls. Soluble TRAIL levels were significantly lower in PDR patients than that in nondiabetic control, whereas soluble RANKL levels did not differ significantly. RANKL, RANK, and TRAIL were expressed in vascular endothelial cells, myofibroblasts, and CD45-expressing leukocytes in PDR epiretinal membranes. CONCLUSIONS Dysregulated expression of OPG/RANKL/RANK pathway and TRAIL might be related to inflammation and angiogenesis in PDR.
Collapse
Affiliation(s)
- Ahmed M Abu El-Asrar
- a Department of Ophthalmology , College of Medicine, King Saud University , Riyadh , Saudi Arabia.,b Dr. Nasser Al-Rashid Research Chair in Ophthalmology, College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Ajmal Ahmad
- a Department of Ophthalmology , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Kaiser Alam
- a Department of Ophthalmology , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Emilie Bittoun
- c Laboratory of Histochemistry and Cytochemistry, Department of Pathology, University of Leuven, KU Leuven , Leuven , Belgium
| | | | - Ghulam Mohammad
- a Department of Ophthalmology , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Ahmed Mousa
- a Department of Ophthalmology , College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | - Gert De Hertogh
- c Laboratory of Histochemistry and Cytochemistry, Department of Pathology, University of Leuven, KU Leuven , Leuven , Belgium
| | - Ghislain Opdenakker
- d Rega Institute for Medical Research, Department of Microbiology and Immunology , University of Leuven, KU Leuven , Leuven , Belgium
| |
Collapse
|
30
|
Bernardi S, Toffoli B, Bossi F, Candido R, Stenner E, Carretta R, Barbone F, Fabris B. Circulating osteoprotegerin is associated with chronic kidney disease in hypertensive patients. BMC Nephrol 2017; 18:219. [PMID: 28683789 PMCID: PMC5500921 DOI: 10.1186/s12882-017-0625-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/19/2017] [Indexed: 12/26/2022] Open
Abstract
Background Osteoprotegerin (OPG) is a glycoprotein that plays an important regulatory role in the skeletal, vascular, and immune system. It has been shown that OPG predicts chronic kidney disease (CKD) in diabetic patients. We hypothesized that OPG could be a risk marker of CKD development also in non-diabetic hypertensive patients. Methods A case-control study was carried out to measure circulating OPG levels in 42 hypertensive patients with CKD and in 141 hypertensive patients without CKD. A potential relationship between OPG and the presence of CKD was investigated and a receiver-operating characteristic (ROC) curve was designed thereafter to identify a cut-off value of OPG that best explained the presence of CKD. Secondly, to evaluate whether OPG increase could affect the kidney, 18 C57BL/6J mice were randomized to be treated with saline or recombinant OPG every 3 weeks for 12 weeks. Results Circulating OPG levels were significantly higher in hypertensive patients with CKD, and there was a significant inverse association between OPG and renal function, that was independent from other variables. ROC analysis showed that OPG levels had a high statistically predictive value on CKD in hypertensive patients, which was greater than that of hypertension. The OPG best cut-off value associated with CKD was 1109.19 ng/L. In the experimental study, OPG delivery significantly increased the gene expression of pro-inflammatory and pro-fibrotic mediators, as well as the glomerular nitrosylation of proteins. Conclusions This study shows that OPG is associated with CKD in hypertensive patients, where it might have a higher predictive value than that of hypertension for CKD development. Secondly, we found that OPG delivery significantly increased the expression of molecular pathways involved in kidney damage. Further longitudinal studies are needed not only to evaluate whether OPG predicts CKD development but also to clarify whether OPG should be considered a risk factor for CKD. Electronic supplementary material The online version of this article (doi:10.1186/s12882-017-0625-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stella Bernardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, Trieste, 34100, Italy. .,Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Strada di Fiume, Trieste, 34100, Italy.
| | - Barbara Toffoli
- IRCCS Burlo Garofolo, Via dell'Istria, Trieste, 34100, Italy
| | - Fleur Bossi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, Trieste, 34100, Italy
| | - Riccardo Candido
- Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Strada di Fiume, Trieste, 34100, Italy
| | - Elisabetta Stenner
- Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Strada di Fiume, Trieste, 34100, Italy
| | - Renzo Carretta
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, Trieste, 34100, Italy.,Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Strada di Fiume, Trieste, 34100, Italy
| | - Fabio Barbone
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, Trieste, 34100, Italy.,IRCCS Burlo Garofolo, Via dell'Istria, Trieste, 34100, Italy
| | - Bruno Fabris
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, Trieste, 34100, Italy.,Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Strada di Fiume, Trieste, 34100, Italy
| |
Collapse
|
31
|
Zhao H, Cao Y, Chen H, Xu W, Sun X, Pan X. The association between OPG rs3102735 gene polymorphism, microembolic signal and stroke severity in acute ischemic stroke patients. Gene 2017; 613:25-29. [DOI: 10.1016/j.gene.2017.02.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/24/2017] [Indexed: 12/27/2022]
|
32
|
Morisawa T, Nakagomi A, Kohashi K, Kusama Y, Shimizu W. Serum Tartrate-resistant Acid Phosphatase-5b Levels are Associated with the Severity and Extent of Coronary Atherosclerosis in Patients with Coronary Artery Disease. J Atheroscler Thromb 2017; 24:1058-1068. [PMID: 28428481 PMCID: PMC5656768 DOI: 10.5551/jat.39339] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aims: Tartrate-resistant acid phosphatase (TRACP)-5b and osteoprotegerin (OPG) are specific and sensitive markers of bone resorption in patients with rheumatoid arthritis (RA) and chronic kidney disease (CKD). The TRACP-5b level is associated with the severity of RA and CKD, while the OPG level is associated with the severity of coronary atherosclerosis and calcification, and can predict a poor outcome in patients with coronary artery disease (CAD). However, the impact of TRACP-5b on coronary atherosclerosis in CAD patients remains unclear. Methods: A total of 71 CAD patients (57 men, 14 women; mean age: 69.0 ± 9.7 years) and 28 age- and gender-matched healthy subjects were investigated. The number of diseased vessels (a marker of the severity of coronary atherosclerosis) and the Gensini score (a marker of the extent of coronary atherosclerosis), as well as the OPG and TRACP-5b levels were measured in CAD patients. The TRACP-5b levels were classified into quartiles. Results: The TRACP-5b levels were significantly higher in CAD patients than in healthy subjects. Patients with higher TRACP-5b levels had higher OPG levels and Gensini scores than those with lower TRACP-5b levels. Higher TRACP-5b levels were associated with an increased number of diseased vessels. A multivariate linear regression analysis showed that the OPG level and the number of diseased vessels or the Gensini score were significantly and independently associated with the TRACP-5b level. Conclusions: These data indicate that the TRACP-5b level is significantly associated with the OPG level and with the severity and extent of coronary atherosclerosis in CAD patients.
Collapse
Affiliation(s)
- Taichirou Morisawa
- Department of Internal Medicine and Cardiology, Tama-Nagayama Hospital, Nippon Medical School
| | - Akihiro Nakagomi
- Department of Internal Medicine and Cardiology, Tama-Nagayama Hospital, Nippon Medical School
| | - Keiichi Kohashi
- Department of Internal Medicine and Cardiology, Tama-Nagayama Hospital, Nippon Medical School
| | - Yoshiki Kusama
- Department of Internal Medicine and Cardiology, Tama-Nagayama Hospital, Nippon Medical School
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School
| |
Collapse
|
33
|
Liu FQ, Liu SQ, Zhang Y, Wang Y, Chu C, Wang D, Pan S, Wang JK, Yu Q, Mu JJ. Effects of Salt Loading on Plasma Osteoprotegerin Levels and Protective Role of Potassium Supplement in Normotensive Subjects. Circ J 2016; 81:77-81. [PMID: 27867157 DOI: 10.1253/circj.cj-16-0756] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Excess dietary salt is strongly correlated with cardiovascular disease, morbidity, and mortality. Conversely, potassium likely elicits favorable effects on cardiovascular disorders. In epidemiological studies, increased plasma osteoprotegerin (OPG) concentrations are associated with atherosclerosis and vascular deaths. Our study was designed to examine the effects of salt intake and potassium supplementation on plasma OPG levels in normotensive subjects. METHODS AND RESULTS The 18 normotensive subjects were selected from a rural community in China. They were sequentially maintained on low-salt diet for 7 days (3 g/day, NaCl), high-salt diet for 7 days (18 g/day), and high-salt diet with potassium supplementation for 7 days (18 g/day of NaCl+4.5 g/day of KCl). High-salt intake enhanced plasma OPG levels (252.7±13.9 vs. 293.4±16.1 pg/mL). This phenomenon was abolished through potassium supplementation (293.4±16.1 vs. 235.1±11.3 pg/mL). Further analyses revealed that the OPG concentration positively correlated with 24-h urinary sodium excretion (r=0.497, P<0.01). By contrast, OPG concentration negatively correlated with 24-h urinary potassium excretion (r=0.594, P<0.01). CONCLUSIONS Salt loading can enhance the production of circulating OPG. Potassium supplementation can reverse the effects of excessive OPG. Our study results may improve our understanding of the roles of salt and potassium in the risk of cardiovascular disorders.
Collapse
Affiliation(s)
- Fu-Qiang Liu
- Cardiovascular Department, Shaanxi Provincial People's Hospital
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Vassalle C, Mazzone A. Bone loss and vascular calcification: A bi-directional interplay? Vascul Pharmacol 2016; 86:77-86. [DOI: 10.1016/j.vph.2016.07.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 06/22/2016] [Accepted: 07/01/2016] [Indexed: 02/07/2023]
|
35
|
Han HH, Kim BG, Lee JH, Kang S, Kim JE, Cho NH. Angiopoietin-2 promotes ER+ breast cancer cell survival in bone marrow niche. Endocr Relat Cancer 2016; 23:609-23. [PMID: 27353038 PMCID: PMC5064757 DOI: 10.1530/erc-16-0086] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022]
Abstract
In estrogen receptor-positive (ER+) breast cancer, it is recognized that metastases may develop after a long period of dormancy. Bone marrow (BM) vascular niche is where the dormant tumor cells are most likely to reside. So far, it is not fully understood why the dormant tumor cells become proliferative and eventually generate tumor. We hypothesized that therapeutic or menopause-related estrogen depletion may be the switch behind dormant ER+ tumor cell awakening in BM. We utilized an existing experimental model of BM endothelial niche that can simulate ER+ tumor cell dormancy to test our hypothesis. In results, estrogen depletion paradoxically promoted ER+ tumor cell proliferation in the BM endothelial niche, and their molecular phenotype shifted from dormant to awaken. Following estrogen depletion, the BM niche cells produced angiopoietin-2 (ANGPT2), which destabilized niche endothelium by interfering ANGPT1/Tie2 signaling, and promoted ER+ tumor cell survival under estrogen deficiency via cell surface integrin &1. Knockdown of ANGPT2 completely negated ER+ tumor cell awakening in the niche. Furthermore, ANGPT2 expression in ER+ tumor human samples was associated with increased risk of distant metastasis only in those who underwent adjuvant estrogen depletion therapy, not in those who did not undergo adjuvant therapy. In conclusion, we demonstrate that ANGPT2 signaling activated after estrogen depletion paradoxically triggers ER+ tumor cell awakening from dormancy in their BM niche, partly indirectly via endothelial Tie2 receptor and partly directly via tumor cell surface integrin &1.
Collapse
Affiliation(s)
- Hyun Ho Han
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of Medicine, Seoul, South Korea Department of PathologyYonsei University College of Medicine, Seoul, South Korea
| | - Baek Gil Kim
- Department of PathologyYonsei University College of Medicine, Seoul, South Korea
| | - Joo Hyun Lee
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of Medicine, Seoul, South Korea
| | - Suki Kang
- Department of PathologyYonsei University College of Medicine, Seoul, South Korea
| | - Ji Eun Kim
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of Medicine, Seoul, South Korea
| | - Nam Hoon Cho
- Brain Korea 21 Plus Project for Medical ScienceYonsei University College of Medicine, Seoul, South Korea Department of PathologyYonsei University College of Medicine, Seoul, South Korea Severance Biomedical Science Institute (SBSI)Yonsei University College of Medicine, Seoul, South Korea Global 5-5-10 System BiologyYonsei University, Seoul, South Korea
| |
Collapse
|
36
|
Bernardi S, Bossi F, Toffoli B, Fabris B. Roles and Clinical Applications of OPG and TRAIL as Biomarkers in Cardiovascular Disease. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1752854. [PMID: 27200369 PMCID: PMC4856888 DOI: 10.1155/2016/1752854] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/28/2016] [Accepted: 04/05/2016] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases (CVD) remain the major cause of death and premature disability in Western societies. Assessing the risk of CVD is an important aspect in clinical decision-making. Among the growing number of molecules that are studied for their potential utility as CVD biomarkers, a lot of attention has been focused on osteoprotegerin (OPG) and its ligands, which are receptor activator of nuclear factor κB ligand (RANKL) and TNF-related apoptosis-inducing ligand. Based on the existing literature and on our experience in this field, here we review what the possible roles of OPG and TRAIL in CVD are and their potential utility as CVD biomarkers.
Collapse
Affiliation(s)
- Stella Bernardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| | - Fleur Bossi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| | - Barbara Toffoli
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| | - Bruno Fabris
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34149 Trieste, Italy
| |
Collapse
|
37
|
Wnt11 Gene Therapy with Adeno-associated Virus 9 Improves Recovery from Myocardial Infarction by Modulating the Inflammatory Response. Sci Rep 2016; 6:21705. [PMID: 26882996 PMCID: PMC4756373 DOI: 10.1038/srep21705] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/29/2016] [Indexed: 12/26/2022] Open
Abstract
Acute myocardial infarction induces activation of the acute phase response and infiltration of leukocytes to the infarcted area. Moreover, myocardium that is remote from ischemic area also becomes inflamed. Inflammatory reaction clears dead cells and matrix debris, while prolongation or expansion of the inflammatory response results in dysfunction following myocardial infarction. Wnt glycolipoproteins are best characterized as regulators of embryonic development. Recently several reports suggest that they also contribute to the inflammatory response in adult animals. However, the effects of Wnt proteins on myocardial infarction have not been explored. Here we show that Wnt11 expression leads to significant improvements of survival and cardiac function by suppressing infiltration of multiple kinds of inflammatory cells in infarcted heart. Wnt11 protein suppresses gene expression of inflammatory cytokines through the modulation of NF-κB in vitro. These results reveal a novel function of Wnt11 in the regulation of inflammatory response and provide a rationale for the use of Wnt11 to manipulate human diseases that are mediated by inflammation.
Collapse
|
38
|
O'Regan N, Moxon C, Gegenbauer K, O'Sullivan JM, Chion A, Smith OP, Preston RJS, Brophy TM, Craig AG, O'Donnell JS. Marked elevation in plasma osteoprotegerin constitutes an early and consistent feature of cerebral malaria. Thromb Haemost 2016; 115:773-80. [PMID: 26766771 PMCID: PMC4990170 DOI: 10.1160/th15-10-0796] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/22/2015] [Indexed: 11/21/2022]
Abstract
Adherence of infected erythrocytes to vascular endothelium causes acute endothelial cell (EC) activation during Plasmodium falciparum infection. Consequently, proteins stored in Weibel-Palade (WP) bodies within EC are secreted into the plasma. Osteoprotegerin (OPG) binds to VWF and consequently is stored within WP bodies. Given the critical role of EC activation in the pathogenesis of severe malaria, we investigated plasma OPG levels in children with P. falciparum malaria. At presentation, plasma OPG levels were significantly elevated in children with cerebral malaria (CM) compared to healthy controls (means 16.0 vs 0.8 ng/ml; p<0.01). Importantly, OPG levels were also significantly higher in children with CM who had a fatal outcome, compared to children with CM who survived. Finally, in children with CM, plasma OPG levels correlated with other established prognostic indices (including plasma lactate levels and peripheral parasite density). To further investigate the relationship between severe malaria and OPG, we utilised a murine model of experimental CM in which C57BL/6J mice were infected with P. berghei ANKA. Interestingly, plasma OPG levels were increased 4.6 fold within 24 hours following P. berghei inoculation. This early marked elevation in OPG levels was observed before any objective clinical signs were apparent, and preceded the development of peripheral blood parasitaemia. As the mice became increasingly unwell, plasma OPG levels progressively increased. Collectively, these data suggest that OPG constitutes a novel biomarker with prognostic significance in patients with severe malaria. In addition, further studies are required to determine whether OPG plays a role in modulating malaria pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - James S O'Donnell
- Prof. James O'Donnell, Haemostasis Research Group, Institute of Molecular Medicine, Trinity College, Dublin, Ireland, Tel.: +353 1 416 2141, Fax: +353 1 410 3570, E-mail:
| |
Collapse
|
39
|
Le CTK, Laidlaw G, Morehouse CA, Naiman B, Brohawn P, Mustelin T, Connor JR, McDonald DM. Synergistic actions of blocking angiopoietin-2 and tumor necrosis factor-α in suppressing remodeling of blood vessels and lymphatics in airway inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2949-68. [PMID: 26348576 DOI: 10.1016/j.ajpath.2015.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 06/22/2015] [Accepted: 07/23/2015] [Indexed: 12/20/2022]
Abstract
Remodeling of blood vessels and lymphatics are prominent features of sustained inflammation. Angiopoietin-2 (Ang2)/Tie2 receptor signaling and tumor necrosis factor-α (TNF)/TNF receptor signaling are known to contribute to these changes in airway inflammation after Mycoplasma pulmonis infection in mice. We determined whether Ang2 and TNF are both essential for the remodeling on blood vessels and lymphatics, and thereby influence the actions of one another. Their respective contributions to the initial stage of vascular remodeling and sprouting lymphangiogenesis were examined by comparing the effects of function-blocking antibodies to Ang2 or TNF, given individually or together during the first week after infection. As indices of efficacy, vascular enlargement, endothelial leakiness, venular marker expression, pericyte changes, and lymphatic vessel sprouting were assessed. Inhibition of Ang2 or TNF alone reduced the remodeling of blood vessels and lymphatics, but inhibition of both together completely prevented these changes. Genome-wide analysis of changes in gene expression revealed synergistic actions of the antibody combination over a broad range of genes and signaling pathways involved in inflammatory responses. These findings demonstrate that Ang2 and TNF are essential and synergistic drivers of remodeling of blood vessels and lymphatics during the initial stage of inflammation after infection. Inhibition of Ang2 and TNF together results in widespread suppression of the inflammatory response.
Collapse
Affiliation(s)
- Catherine T K Le
- Department of Anatomy, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California; University of California San Francisco Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Grace Laidlaw
- Department of Anatomy, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California; University of California San Francisco Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | | | - Brian Naiman
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; MedImmune LLC, Gaithersburg, Maryland
| | | | | | | | - Donald M McDonald
- Department of Anatomy, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California; University of California San Francisco Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California.
| |
Collapse
|
40
|
Esteghamati A, Aflatoonian M, Rad MV, Mazaheri T, Mousavizadeh M, Nakhjavani M, Noshad S. Association of osteoprotegerin with peripheral artery disease in patients with type 2 diabetes. Arch Cardiovasc Dis 2015; 108:412-9. [PMID: 26184866 DOI: 10.1016/j.acvd.2015.01.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 11/30/2014] [Accepted: 01/28/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Osteoprotegerin plays a critical role in the pathogenesis of atherosclerosis. Elevated osteoprotegerin concentrations have been reported in microvascular complications of diabetes. Patients with diabetes are at increased risk of macrovascular complications, particularly peripheral artery disease (PAD). AIM To investigate the association between osteoprotegerin concentration and PAD in diabetes. METHODS In a cross-sectional setting, patients with type 2 diabetes for>5 years and no apparent diabetic foot ulcer were recruited. Patients underwent colour Doppler ultrasonography of lower limbs and were designated PAD+ if arterial narrowing was detected. Ankle-brachial index (ABI) was measured. Serum osteoprotegerin concentrations were determined. RESULTS Ninety-eight patients (47 PAD+, 51 PAD-) were recruited. Osteoprotegerin concentrations (median [interquartile range]) were significantly higher in PAD+ versus PAD- patients (0.80 [0.50-1.95] ng/mL vs 0.30 [0.25-0.40] ng/mL; P<0.001). In logistic regression, log-osteoprotegerin was a predictor of PAD in univariate and multivariable analyses. In the final multivariable model, adjusting for age, sex, body mass index, smoking, hypertension, glycaemic control, lipid profile, renal function and C-reactive protein, one standard deviation increase in log-osteoprotegerin was associated with a more than twofold increase in the risk of having PAD (odds ratio 2.26, 95% confidence interval 1.50-3.40). In PAD+ patients, osteoprotegerin was a significant predictor of disease severity, determined by ABI and percentage of vessel occlusion in univariate and multivariable models. CONCLUSIONS Osteoprotegerin concentrations are increased in patients with diabetes and PAD. Osteoprotegerin is an independent predictor of the presence and severity of PAD in diabetic patients.
Collapse
Affiliation(s)
- Alireza Esteghamati
- Endocrinology and Metabolism Research Centre (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran.
| | - Maryam Aflatoonian
- Endocrinology and Metabolism Research Centre (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Mona Vahidi Rad
- Endocrinology and Metabolism Research Centre (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Tina Mazaheri
- Endocrinology and Metabolism Research Centre (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Mostafa Mousavizadeh
- Endocrinology and Metabolism Research Centre (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Manouchehr Nakhjavani
- Endocrinology and Metabolism Research Centre (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Sina Noshad
- Endocrinology and Metabolism Research Centre (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| |
Collapse
|
41
|
Osteoprotegerin exposure at different stages of osteoclastogenesis differentially affects osteoclast formation and function. Cytotechnology 2015; 68:1325-35. [PMID: 26044733 DOI: 10.1007/s10616-015-9892-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/23/2015] [Indexed: 10/23/2022] Open
Abstract
This study aimed to investigate the effects of osteoprotegerin (OPG), a decoy receptor for receptor activator for nuclear factor κB ligand (RANKL), during the various stages of osteoclast differentiation, and additionally investigate its effects on osteoclast adhesion and activity. RAW264.7 murine monocytic cells were incubated with macrophage colony-stimulating factor and RANKL for 1, 3, 5, or 7 days, followed by an additional 24-h incubation in the presence or absence of OPG (80 ng/mL). We examined osteoclast differentiation and adhesion capacity using the tartrate-resistant acid phosphatase (TRAP) assay and immunofluorescence microscopy, and additionally examined cell growth in real time using the xCELLigence system. Furthermore, the expression levels of TRAP, RANK, integrin β3, matrix metalloproteinase 9, cathepsin K, carbonic anhydrase II, and vesicular-type H(+)-ATPase A1 were examined using western blotting. OPG exposure on day 1 enhanced the osteoclast growth curve as well as adhesion, and increased RANK and integrin β3 expression. In contrast, exposure to OPG at later time points (days 3-7) inhibited osteoclast differentiation, adhesion structure formation, and protease expression. In conclusion, the biological effects of OPG exposure at the various stages of osteoclast differentiation were varied, and included the enhanced adhesion and survival of preosteoclasts, the block of differentiation from the early to the terminal stages of osteoclastogenesis, and suppression of mature osteoclast activation following OPG exposure during the terminal differentiation stage, suggesting that the effects of OPG exposure differ based on the stage of differentiation.
Collapse
|
42
|
Increased osteoprotegerin predicts poor virological outcome during anticytomegalovirus therapy in solid organ transplant recipients. Transplantation 2015; 99:100-5. [PMID: 24983306 DOI: 10.1097/tp.0000000000000227] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) infection involves interaction between endothelial cells and leukocyte subsets that may promote vascular inflammation and lead to treatment failure in infected individuals. Osteoprotegerin is a marker of vascular and systemic inflammation but has not been investigated in relation to treatment outcome during CMV infection. METHODS We investigated whether circulating levels of osteoprotegerin are related to features of CMV disease and treatment outcomes during CMV infection in 291 solid organ transplant recipients receiving valganciclovir or ganciclovir in an international multicenter trial of CMV disease treatment (the VICTOR study). RESULTS Elevated plasma osteoprotegerin was associated with (i) certain disease characteristics including presence of tissue invasive disease (P<0.05) and increased viral load at baseline (P<0.05), (ii) poor virological outcome at day 49 after anti-CMV therapy, (iii) increased plasma levels of markers of inflammation (pentraxin 3 and C-reactive protein) and endothelial cell activation (von Willebrand factor) both at baseline and during follow-up. CONCLUSION Our finding indicates that elevated osteoprotegerin levels in solid organ transplant recipients with CMV infection may reflect vascular inflammation and is associated with late virological outcome in these patients.
Collapse
|
43
|
Pérez de Ciriza C, Lawrie A, Varo N. Osteoprotegerin in Cardiometabolic Disorders. Int J Endocrinol 2015; 2015:564934. [PMID: 26078757 PMCID: PMC4442310 DOI: 10.1155/2015/564934] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 04/20/2015] [Accepted: 04/20/2015] [Indexed: 02/06/2023] Open
Abstract
Osteoprotegerin (OPG), a glycoprotein traditionally implicated in bone remodelling, has been recently related to cardiovascular disease (CVD). Human studies show a positive relationship between circulating OPG, vascular damage, and CVD, and as such OPG has emerged as a potential biomarker for CVD. This review focuses on the relationship between circulating OPG and different endocrine cardiometabolic alterations such as type 1 and 2 diabetes. The association of OPG with diabetic complications (neuropathy, nephropathy, or retinopathy) as well as with atherosclerosis, coronary artery calcification, morbidity, and mortality is pointed out. Moreover, OPG modulation by different treatments is also established. Besides, other associated diseases such as obesity, hypertension, and metabolic syndrome, which are known cardiovascular risk factors, are also considered.
Collapse
Affiliation(s)
- C. Pérez de Ciriza
- Department of Clinical Chemistry, Clínica Universidad de Navarra, Avenida Pío XII 36, 31008 Pamplona, Spain
| | - A. Lawrie
- Department of Cardiovascular Science, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - N. Varo
- Department of Clinical Chemistry, Clínica Universidad de Navarra, Avenida Pío XII 36, 31008 Pamplona, Spain
- *N. Varo:
| |
Collapse
|
44
|
López-Mejias R, Ubilla B, Genre F, Corrales A, Hernández JL, Ferraz-Amaro I, Tsang L, Llorca J, Blanco R, González-Juanatey C, Gonzalez-Gay MA, Dessein PH. Osteoprotegerin concentrations relate independently to established cardiovascular disease in rheumatoid arthritis. J Rheumatol 2014; 42:39-45. [PMID: 25362655 DOI: 10.3899/jrheum.140690] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE We determined whether osteoprotegerin (OPG) concentrations are associated with established cardiovascular disease (CVD) among patients with rheumatoid arthritis (RA). METHODS OPG concentrations were measured by ELISA in 151 patients with RA (54 with CVD) and 62 age-matched control subjects without CVD. Established CVD was composed of documented ischemic heart disease, cerebrovascular disease, and peripheral artery disease. RESULTS In patients with RA, age, body mass index (BMI), rheumatoid factor (RF) positivity, anticyclic citrullinated peptide (anti-CCP) antibody positivity, and joint erosion status were associated with OPG concentrations [partial R (p) = 0.175 (0.03), -0.277 (0.0009), 0.323 (< 0.0001), 0.217 (0.008), and 0.159 (0.05), respectively]. Median (interquartile range) OPG concentrations increased from 6.38 (3.46-9.31) to 7.07 (5.04-10.65) and 8.64 (6.00-11.52) ng/ml in controls and patients with RA who had CVD and those who did not, respectively (p = 0.0002). Upon adjustment for age, sex, traditional risk factors, and BMI in mixed regression models, OPG concentrations remained lower in controls compared to patients with RA without CVD (p = 0.05) and in the latter compared to those with CVD (p = 0.03); the association of OPG concentrations with CVD among patients with RA also persisted after additional adjustment for RF and anti-CCP antibody positivity, and erosion status (p = 0.04). CONCLUSION OPG concentrations are associated with disease severity and CVD prevalence in patients with RA. Whether consideration of OPG concentrations can improve CVD risk stratification in RA merits future longitudinal investigation.
Collapse
Affiliation(s)
- Raquel López-Mejias
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - Begoña Ubilla
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - Fernanda Genre
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - Alfonso Corrales
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - José L Hernández
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - Ivan Ferraz-Amaro
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - Linda Tsang
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - Javier Llorca
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - Ricardo Blanco
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - Carlos González-Juanatey
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - Miguel A Gonzalez-Gay
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| | - Patrick H Dessein
- From the Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, and the Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and Fundación Instituto de Investigación Marqués de Valdecilla, and Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), and CIBER Epidemiología y Salud Pública (CIBERESP), Santander; the Rheumatology Division, Hospital Universitario de Canarias, Tenerife; the Cardiology Division, Hospital Lucus Augusti, Lugo, Spain; the Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.P.H. Dessein and M.A. Gonzalez-Gay share senior authorship.R. López-Mejias, PhD; B. Ubilla, BSc; F. Genre, PhD; A. Corrales, MD, PhD; R. Blanco, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV; J.L. Hernández, MD, PhD, Department of Internal Medicine, Hospital Universitario Marques de Valdecilla, University of Cantabria, RETICEF, IFIMAV; I. Ferraz-Amaro, MD, Rheumatology Division, Hospital Universitario de Canarias; L. Tsang; P.H. Dessein, MD, FCP(SA), FRCP(UK), PhD, Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand; J. Llorca, MD, PhD, Department of Epidemiology and Computational Biology, School of Medicine, University of Cantabria, and CIBERESP, IFIMAV; C. Gonzalez-Juanatey, MD, PhD, Cardiology Division, Hospital Lucus Augusti; M.A. Gonzalez-Gay, MD, PhD, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marques de Valdecilla, IFIMAV
| |
Collapse
|
45
|
Abali R, Tasdemir N, Alpsoy S, Tasdemir UG, Guzel S, Yuksel MA, Temel Yuksel I, Yilmaz M. No relationship between osteoprotegerin concentrations and endothelial dysfunction in non-obese women with and without polycystic ovary syndrome. Arch Gynecol Obstet 2014; 291:1075-80. [PMID: 25280574 DOI: 10.1007/s00404-014-3499-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 09/25/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE To investigate the relationships of osteoprotegerin (OPG) concentrations to brachial artery flow-mediated vasodilation (FMD) and the carotid artery intima media thickness (CIMT) in polycystic ovary syndrome (PCOS). METHODS Thirty-seven women with PCOS and 41 controls matched for body mass index (BMI) and age were included in study. The serum OPG concentrations, hormonal and metabolic profiles were measured in women with PCOS and in control group. The CIMT and brachial artery FMD were evaluated in both groups. RESULTS The mean serum concentrations of all hormones were comparable, except LH, which was higher in women with PCOS. Lipid parameters were similar between groups. There were no differences between groups with respect to fasting glucose, 2-h glucose, fasting insulin, HbA1c and HOMA-IR. The mean osteoprotogerin concentrations were higher in PCOS group (11.39 ± 2.29 vs. 10.22 ± 2.25 pmol/L, P = 0.026). The mean CIMT was higher in PCOS group than control group (0.52 ± 0.058 vs. 0.45 ± 0.059 mm, P < 0.01). The mean brachial artery FMD was lower in PCOS group (0.068 ± 0.022 vs. 0.055 ± 0.029, P = 0.017). CONCLUSIONS We found high osteoprotogerin concentrations, increased CIMT and decreased FMD, in women with PCOS. However, there was no correlation between osteoprotegerin and cardiovascular risk markers.
Collapse
Affiliation(s)
- Remzi Abali
- Department of Gynecology and Obstetrics, Faculty of Medicine, Namik Kemal University, 100. YilMah. Barbaros Cad, No: 132, Tekirdag, Turkey,
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Bernardi S, Fabris B, Thomas M, Toffoli B, Tikellis C, Candido R, Catena C, Mulatero P, Barbone F, Radillo O, Zauli G, Secchiero P. Osteoprotegerin increases in metabolic syndrome and promotes adipose tissue proinflammatory changes. Mol Cell Endocrinol 2014; 394:13-20. [PMID: 24998520 DOI: 10.1016/j.mce.2014.06.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 06/08/2014] [Accepted: 06/09/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Inflammation is believed to link obesity to insulin resistance, as in the setting of metabolic syndrome (MetS). Osteoprotegerin (OPG) is a soluble protein that seems to exert proatherogenic and prodiabetogenic effects. This study aims at determining OPG levels in MetS and whether OPG might contribute to MetS development and progression. METHODOLOGY/PRINCIPAL FINDINGS Circulating OPG was measured in 46 patients with MetS and 63 controls, and was found significantly elevated in those with MetS. In addition, circulating and tissue OPG was significantly increased in high-fat diet (HFD) fed C57BL6 mice, which is one of the animal models for the study of MetS. To evaluate the consequences of OPG elevation, we delivered this protein to C57BL6 mice, finding that it promoted systemic and adipose tissue proinflammatory changes in association with metabolic abnormalities. CONCLUSIONS/SIGNIFICANCE These data suggest that OPG may trigger adipose tissue proinflammatory changes in MetS/HFD-induced obesity.
Collapse
Affiliation(s)
- Stella Bernardi
- Department of Morphology, Surgery and Experimental Medicine, LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44100 Ferrara, Italy; Baker IDI, Heart and Diabetes Research Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.
| | - Bruno Fabris
- Department of Medical, Surgical and Health Sciences, University of Trieste, Ospedale di Cattinara, Strada di Fiume 447, 34149 Trieste, Italy.
| | - Merlin Thomas
- Baker IDI, Heart and Diabetes Research Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.
| | - Barbara Toffoli
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34100 Trieste, Italy.
| | - Christos Tikellis
- Baker IDI, Heart and Diabetes Research Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia.
| | | | - Cristiana Catena
- Department of Medical and Biological Sciences, University of Udine, Ospedale Santa Maria della Misericordia, Udine, Italy.
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension, University of Torino, Ospedale San Giovanni Battista, 10126 Torino, Italy.
| | - Fabio Barbone
- Department of Medical, Experimental and Clinical Sciences, University of Udine, Ospedale Santa Maria della Misericordia, Udine, Italy.
| | - Oriano Radillo
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34100 Trieste, Italy.
| | - Giorgio Zauli
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34100 Trieste, Italy.
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine, LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44100 Ferrara, Italy.
| |
Collapse
|
47
|
Guan S, Wang Z, Xin F, Xin H. Wnt5a is associated with the differentiation of bone marrow mesenchymal stem cells in vascular calcification by connecting with different receptors. Mol Med Rep 2014; 10:1985-91. [PMID: 25109262 DOI: 10.3892/mmr.2014.2449] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 05/23/2014] [Indexed: 11/06/2022] Open
Abstract
Vascular calcification significantly affects the health of the elderly. Increasing evidence proved that vascular calcification is an actively regulated osteogenic process. The osteochondrocytic differentiation of mesenchymal stem cells (MSCs) is a significant step of osteogenic processes. The Wnt pathways has been identified as contributing to the regulation of osteogenic mineralization during development and disease. However, it remains unknown whether these MSCs in the vascular calcification differentiate into normal vascular smooth muscle cells (VSMCs) in vivo in order to treat damaged vascular tissue or into calcified VSMCs to aggravate calcification correlated to the Wnt pathways. Thus, it is necessary to analyze the mechanisms of MSC differentiation in detail. In the present study a cell‑cell co‑culturing in vitro system was used to observe MSCs that directly interact with normal or calcified VSMCs during calcification and to investigate the gene expression of the Wnt pathways during the process. Direct co‑cultures were established by seeding two different cell types, VSMCs or calcified VSMCs, or a mixture of both at ratios of 5,000:5,000 cells/1.7 cm2 onto either gelatin‑coated 1.7‑cm2 chamber slides for immunohistochemical analysis or gelatin‑coated 75‑cm2 tissue culture flasks for protein or RNA isolation. Osteoblastic differentiation was evaluated by examining the cell morphology and assessing the activity of alkaline phosphatase in the cell lysates by alkaline phosphatase staining. Additionally, the mRNA expression levels of the genes encoding for proteins involved in the Wnt signaling proteins, Wnt5A, LRP6, Ror2, c‑Jun‑N‑terminal kinase and β‑catenin, were assessed in each group. The present study demonstrated that Wnts are expressed in the progress of differentiation of MSCs during calcification. MSCs can differentiate into different cell phenotypes when there is direct cell‑cell contact with VSMCs or calcified VSMCs, and the Wnt5a/Ror2 signaling pathway may be associated with the determination of differentiation of MSCs in this process.
Collapse
Affiliation(s)
- Siming Guan
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhimin Wang
- Department of Neurology, The First People's Hospital of Taizhou, Taizhou, Zhejiang 318020, P.R. China
| | - Fang Xin
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Huaping Xin
- Department of Neurology, The First People's Hospital of Taizhou, Taizhou, Zhejiang 318020, P.R. China
| |
Collapse
|
48
|
Fuernau G, Poenisch C, Eitel I, de Waha S, Desch S, Schuler G, Adams V, Werdan K, Zeymer U, Thiele H. Growth-differentiation factor 15 and osteoprotegerin in acute myocardial infarction complicated by cardiogenic shock: a biomarker substudy of the IABP-SHOCK II-trial. Eur J Heart Fail 2014; 16:880-7. [DOI: 10.1002/ejhf.117] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/26/2014] [Accepted: 05/02/2014] [Indexed: 01/08/2023] Open
Affiliation(s)
- Georg Fuernau
- Department of Internal Medicine/Cardiology; University of Leipzig-Heart Center; Leipzig Germany
| | - Christian Poenisch
- Department of Internal Medicine/Cardiology; University of Leipzig-Heart Center; Leipzig Germany
| | - Ingo Eitel
- Department of Internal Medicine/Cardiology; University of Leipzig-Heart Center; Leipzig Germany
| | - Suzanne de Waha
- Department of Internal Medicine/Cardiology; University of Leipzig-Heart Center; Leipzig Germany
| | - Steffen Desch
- Department of Internal Medicine/Cardiology; University of Leipzig-Heart Center; Leipzig Germany
| | - Gerhard Schuler
- Department of Internal Medicine/Cardiology; University of Leipzig-Heart Center; Leipzig Germany
| | - Volker Adams
- Department of Internal Medicine/Cardiology; University of Leipzig-Heart Center; Leipzig Germany
| | - Karl Werdan
- Department of Medicine III; University Clinics of the Martin Luther University Halle-Wittenberg; Halle/Saale Germany
| | - Uwe Zeymer
- Medizinische Klinik B, Klinikum Ludwigshafen and Institut für Herzinfarktforschung; Ludwigshafen Germany
| | - Holger Thiele
- Department of Internal Medicine/Cardiology; University of Leipzig-Heart Center; Leipzig Germany
- Medical Clinic II; University of Lübeck; Lübeck Germany
| |
Collapse
|
49
|
Bollmann F, Wu Z, Oelze M, Siuda D, Xia N, Henke J, Daiber A, Li H, Stumpo DJ, Blackshear PJ, Kleinert H, Pautz A. Endothelial dysfunction in tristetraprolin-deficient mice is not caused by enhanced tumor necrosis factor-α expression. J Biol Chem 2014; 289:15653-65. [PMID: 24727475 PMCID: PMC4140920 DOI: 10.1074/jbc.m114.566984] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 04/09/2014] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular events are important co-morbidities in patients with chronic inflammatory diseases like rheumatoid arthritis. Tristetraprolin (TTP) regulates pro-inflammatory processes through mRNA destabilization and therefore TTP-deficient mice (TTP(-/-) mice) develop a chronic inflammation resembling human rheumatoid arthritis. We used this mouse model to evaluate molecular signaling pathways contributing to the enhanced atherosclerotic risk in chronic inflammatory diseases. In the aorta of TTP(-/-) mice we observed elevated mRNA expression of known TTP targets like tumor necrosis factor-α (TNF-α) and macrophage inflammatory protein-1α, as well as of other pro-atherosclerotic mediators, like Calgranulin A, Cathepsin S, and Osteopontin. Independent of cholesterol levels TTP(-/-) mice showed a significant reduction of acetylcholine-induced, nitric oxide-mediated vasorelaxation. The endothelial dysfunction in TTP(-/-) mice was associated with increased levels of reactive oxygen and nitrogen species (RONS), indicating an enhanced nitric oxide inactivation by RONS in the TTP(-/-) animals. The altered RONS generation correlates with increased expression of NADPH oxidase 2 (Nox2) resulting from enhanced Nox2 mRNA stability. Although TNF-α is believed to be a central mediator of inflammation-driven atherosclerosis, genetic inactivation of TNF-α neither improved endothelial function nor normalized Nox2 expression or RONS production in TTP(-/-) animals. Systemic inflammation caused by TTP deficiency leads to endothelial dysfunction. This process is independent of cholesterol and not mediated by TNF-α solely. Thus, other mediators, which need to be identified, contribute to enhanced cardiovascular risk in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Franziska Bollmann
- From the Department of Pharmacology, Center for Thrombosis and Hemostasis, and
| | | | - Matthias Oelze
- 2nd Medical Clinic, Molecular Cardiology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany and
| | - Daniel Siuda
- From the Department of Pharmacology, Center for Thrombosis and Hemostasis, and
| | - Ning Xia
- From the Department of Pharmacology
| | | | - Andreas Daiber
- 2nd Medical Clinic, Molecular Cardiology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany and
| | - Huige Li
- From the Department of Pharmacology
| | - Deborah J Stumpo
- the Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | - Perry J Blackshear
- the Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709
| | | | | |
Collapse
|
50
|
Adipokines, biomarkers of endothelial activation, and metabolic syndrome in patients with ankylosing spondylitis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:860651. [PMID: 24757680 PMCID: PMC3976882 DOI: 10.1155/2014/860651] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/13/2014] [Indexed: 12/11/2022]
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory rheumatic disease associated with accelerated atherosclerosis and increased risk of cardiovascular (CV) disease. AS patients also display a high prevalence of features clustered under the name of metabolic syndrome (MeS). Anti-TNF-α therapy was found to be effective to treat AS patients by suppressing inflammation and also improving endothelial function. Previously, it was demonstrated that a short infusion of anti-TNF-α monoclonal antibodyinfliximab induced a rapid and dramatic reduction in serum insulin levels and insulin resistance along with a rapid improvement of insulin sensitivity in nondiabetic AS patients. The role of adipokines, MeS-related biomarkers and biomarkers of endothelial cell activation and inflammation seem to be relevant in different chronic inflammatory diseases. However, its implication in AS has not been fully established. Therefore, in this review we summarize the recent advances in the study of the involvement of these molecules in CV disease or MeS in AS. The assessment of adipokines and biomarkers of endothelial cell activation and MeS may be of potential relevance in the stratification of the CV risk of patients with AS.
Collapse
|