1
|
Mo M, Yin L, Wang T, Lv Z, Guo Y, Shen J, Zhang H, Liu N, Wang Q, Huang S, Huang H. Associations of essential metals with the risk of aortic arch calcification: a cross-sectional study in a mid-aged and older population of Shenzhen, China. MedComm (Beijing) 2024; 5:e533. [PMID: 38745853 PMCID: PMC11091022 DOI: 10.1002/mco2.533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/10/2024] [Accepted: 03/12/2024] [Indexed: 05/16/2024] Open
Abstract
Vascular calcification is a strong predictor of cardiovascular events. Essential metals play critical roles in maintaining human health. However, the association of essential metal levels with risk of aortic arch calcification (AoAC) remains unclear. We measured the plasma concentrations of nine essential metals in a cross-sectional population and evaluated their individual and combined effects on AoAC risk using multiple statistical methods. We also explored the mediating role of fasting glucose. In the logistic regression model, higher quartiles of magnesium and copper were associated with the decreased AoAC risk, while higher quartile of manganese was associated with higher AoAC risk. The least absolute shrinkage and selection operator penalized regression analysis identified magnesium, manganese, calcium, cobalt, and copper as key metals associated with AoAC risk. The weighted quantile sum regression suggested a combined effect of metal mixture. A linear and positive dose-response relationship was found between manganese and AoAC in males. Moreover, blood glucose might mediate a proportion of 9.38% of the association between manganese exposure and AoAC risk. In summary, five essential metal levels were associated with AoAC and showed combined effect. Fasting glucose might play a significant role in mediating manganese exposure-associated AoAC risk.
Collapse
Affiliation(s)
- Mingxing Mo
- Department of CardiologyJoint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseasesthe Eighth Affiliated Hospital of Sun Yat‐sen UniversityShenzhenChina
| | - Li Yin
- Department of CardiologyJoint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseasesthe Eighth Affiliated Hospital of Sun Yat‐sen UniversityShenzhenChina
| | - Tian Wang
- School of Public HealthShenzhen University Medical SchoolShenzhen UniversityShenzhenGuangdongChina
- Department of Central LaboratoryShenzhen Center for Disease control and PreventionShenzhenChina
| | - Ziquan Lv
- Department of Central LaboratoryShenzhen Center for Disease control and PreventionShenzhenChina
| | - Yadi Guo
- Department of CardiologyJoint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseasesthe Eighth Affiliated Hospital of Sun Yat‐sen UniversityShenzhenChina
| | - Jiangang Shen
- School of Chinese MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong SARChina
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong SARChina
| | - Huanji Zhang
- Department of CardiologyJoint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseasesthe Eighth Affiliated Hospital of Sun Yat‐sen UniversityShenzhenChina
| | - Ning Liu
- Department of Central LaboratoryShenzhen Center for Disease control and PreventionShenzhenChina
| | - Qiuling Wang
- Department of Central LaboratoryShenzhen Center for Disease control and PreventionShenzhenChina
| | - Suli Huang
- School of Public HealthShenzhen University Medical SchoolShenzhen UniversityShenzhenGuangdongChina
- Department of Central LaboratoryShenzhen Center for Disease control and PreventionShenzhenChina
| | - Hui Huang
- Department of CardiologyJoint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseasesthe Eighth Affiliated Hospital of Sun Yat‐sen UniversityShenzhenChina
| |
Collapse
|
2
|
Sun X, Zheng Y, Xie L, Zhou Y, Liu R, Ma Y, Zhao M, Liu Y. Autophagy reduces aortic calcification in diabetic mice by reducing matrix vesicle body-mediated IL-1β release. Exp Cell Res 2023; 432:113803. [PMID: 37774764 DOI: 10.1016/j.yexcr.2023.113803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Vascular calcification (VC) is a common pathological process of cardiovascular disease that occurs in patients with type 2 diabetes mellitus (T2DM). However, the molecular basis of VC progression remains unknown. A GEO dataset (GSE146638) was analyzed to show that microbodies and IL-1β may play important roles in the pathophysiology of VC. The release of matrix vesicle bodies (MVBs) and IL-1β and the colocalization of IL-1β with MVBs or autophagosomes were studied by immunofluorescence in an in vivo diabetes mouse model with aortic calcification and an in vitro high glucose cell calcification model. MVB numbers, IL-1β levels and autophagy were increased in calcified mouse aortas and calcified vascular smooth muscle cells (VSMCs). IL-1β colocalized with MVBs and autophagosomes. The MVBs from calcified VSMCs induced the calcification of normal recipient VSMCs, and this effect was alleviated by silencing IL-1β. The autophagy inducer rapamycin reduced IL-1β expression and calcification in VSMCs, while these processes were induced by the autophagy inhibitor chloroquine. In conclusion, our results suggested that MVBs could carry IL-1β out of cells and induce VC in normal VSMCs, and these processes could be counteracted by autophagy. These results suggested that MVB-mediated IL-1β release may be an effective target for treating vascular calcification.
Collapse
Affiliation(s)
- Xiaolei Sun
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Laboratory of Nucleic Acids in Medicine for National High-Level Talents, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China; Cardiovascular and Metabolic Diseases Key Laboratory of Sichuan, Luzhou, 646000, China.
| | - Yang Zheng
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Vascular and Interventional, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Linzhuo Xie
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yuanqun Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China; State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Runyu Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yarong Ma
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ming Zhao
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China.
| | - Yong Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
3
|
Ho LC, Chen YH, Wu TY, Kao LZ, Hung SY, Liou HH, Chen PC, Tsai PJ, Lin HK, Lee YC, Wang HH, Tsai YS. Phosphate burden induces vascular calcification through a NLRP3-caspase-1-mediated pyroptotic pathway. Life Sci 2023; 332:122123. [PMID: 37742736 DOI: 10.1016/j.lfs.2023.122123] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/01/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
AIMS The aim of this study is to clarify the role of NLRP3 inflammasome in phosphate burden-induced vascular smooth muscle cell (VSMC) calcification. MAIN METHODS VSMC calcification was induced using a high concentration of inorganic phosphate. After pharmacological inhibition or genetic silencing of the NLRP3 inflammasome, pyroptosis, or potassium efflux, the cells were examined by RT-qPCR, immunofluorescence, and western blotting to identify the NLRP3-mediated pathway for VSMC calcification. KEY FINDINGS Calcified VSMCs with α-smooth muscle actin (α-SMA) disarray presented features of pyroptosis, including caspase-1 maturation, cleaved gasdermin D (GSDMD), and a high supernatant level of lactate dehydrogenase A. Pharmacological inhibitions of caspase-1 and pyroptosis attenuated VSMC calcification, whereas interleukin-1β receptor antagonism did not. Unlike canonical NLRP3 activation, osteogenic VSMCs did not upregulate NLRP3 expression. However, NLRP3 genetic silencing or inhibitions, which targets different domains of the NLRP3 protein, could ameliorate VSMC calcification by aborting caspase-1 and GSDMD activation. Furthermore, potassium efflux through the inward-rectifier potassium channel, and not through the P2X7 receptor, triggered NLRP3 inflammasome activation and VSMC calcification. SIGNIFICANCE In the present study, we identified a potassium efflux-triggered NLRP3-caspase-1-mediated pyroptotic pathway for VSMC calcification that is unique and different from the canonical NLRP3 inflammasome activation. Therefore, targeting this pathway may serve as a novel therapeutic strategy for vascular calcification.
Collapse
Affiliation(s)
- Li-Chun Ho
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Division of General Medicine, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.
| | - Yu-Hsin Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Yun Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ling-Zhen Kao
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Yuan Hung
- Division of Nephrology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Hung-Hsiang Liou
- Division of Nephrology, Department of Internal Medicine, Hsin-Jen Hospital, New Taipei City, Taiwan
| | - Pei-Chun Chen
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Yi-Che Lee
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Hsi-Hao Wang
- Division of Nephrology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yau-Sheng Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA; Clinical Medicine Research Center, National Cheng Kung University Hospital, Tainan, Taiwan.
| |
Collapse
|
4
|
Ding N, Lv Y, Su H, Wang Z, Kong X, Zhen J, Lv Z, Wang R. Vascular calcification in CKD: New insights into its mechanisms. J Cell Physiol 2023; 238:1160-1182. [PMID: 37269534 DOI: 10.1002/jcp.31021] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/28/2023] [Indexed: 06/05/2023]
Abstract
Vascular calcification (VC) is a common complication of chronic kidney disease (CKD) and contributes to an increased risk of cardiovascular morbidity and mortality. However, effective therapies are still unavailable at present. It has been well established that VC associated with CKD is not a passive process of calcium phosphate deposition, but an actively regulated and cell-mediated process that shares many similarities with bone formation. Additionally, numerous studies have suggested that CKD patients have specific risk factors and contributors to the development of VC, such as hyperphosphatemia, uremic toxins, oxidative stress and inflammation. Although research efforts in the past decade have greatly improved our knowledge of the multiple factors and mechanisms involved in CKD-related VC, many questions remain unanswered. Moreover, studies from the past decade have demonstrated that epigenetic modifications abnormalities, such as DNA methylation, histone modifications and noncoding RNAs, play an important role in the regulation of VC. This review seeks to provide an overview of the pathophysiological and molecular mechanisms of VC associated with CKD, mainly focusing on the involvement of epigenetic modifications in the initiation and progression of uremic VC, with the aim to develop promising therapies for CKD-related cardiovascular events in the future.
Collapse
Affiliation(s)
- Nannan Ding
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yaodong Lv
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Hong Su
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ziyang Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xianglei Kong
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Junhui Zhen
- Department of Pathology, Shandong University, Jinan, China
| | - Zhimei Lv
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
5
|
Bessueille L, Kawtharany L, Quillard T, Goettsch C, Briolay A, Taraconat N, Balayssac S, Gilard V, Mebarek S, Peyruchaud O, Duboeuf F, Bouillot C, Pinkerton A, Mechtouff L, Buchet R, Hamade E, Zibara K, Fonta C, Canet-Soulas E, Millan JL, Magne D. Inhibition of alkaline phosphatase impairs dyslipidemia and protects mice from atherosclerosis. Transl Res 2023; 251:2-13. [PMID: 35724933 DOI: 10.1016/j.trsl.2022.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022]
Abstract
Calcium accumulation in atherosclerotic plaques predicts cardiovascular mortality, but the mechanisms responsible for plaque calcification and how calcification impacts plaque stability remain debated. Tissue-nonspecific alkaline phosphatase (TNAP) recently emerged as a promising therapeutic target to block cardiovascular calcification. In this study, we sought to investigate the effect of the recently developed TNAP inhibitor SBI-425 on atherosclerosis plaque calcification and progression. TNAP levels were investigated in ApoE-deficient mice fed a high-fat diet from 10 weeks of age and in plaques from the human ECLAGEN biocollection (101 calcified and 14 non-calcified carotid plaques). TNAP was inhibited in mice using SBI-425 administered from 10 to 25 weeks of age, and in human vascular smooth muscle cells (VSMCs) with MLS-0038949. Plaque calcification was imaged in vivo with 18F-NaF-PET/CT, ex vivo with osteosense, and in vitro with alizarin red. Bone architecture was determined with µCT. TNAP activation preceded and predicted calcification in human and mouse plaques, and TNAP inhibition prevented calcification in human VSMCs and in ApoE-deficient mice. More unexpectedly, TNAP inhibition reduced the blood levels of cholesterol and triglycerides, and protected mice from atherosclerosis, without impacting the skeletal architecture. Metabolomics analysis of liver extracts identified phosphocholine as a substrate of liver TNAP, who's decreased dephosphorylation upon TNAP inhibition likely reduced the release of cholesterol and triglycerides into the blood. Systemic inhibition of TNAP protects from atherosclerosis, by ameliorating dyslipidemia, and preventing plaque calcification.
Collapse
Affiliation(s)
- Laurence Bessueille
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Lynn Kawtharany
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Thibaut Quillard
- CNRS, INSERM, l'institut du thorax, Nantes Université, Nantes, France
| | - Claudia Goettsch
- Department of Internal Medicine I, Cardiology, Medical Faculty, RWTH Aachen University, Aachen Germany
| | - Anne Briolay
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Nirina Taraconat
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III, Paul Sabatier, France
| | - Stéphane Balayssac
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III, Paul Sabatier, France
| | - Véronique Gilard
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III, Paul Sabatier, France
| | - Saida Mebarek
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | | | | | | | | | - Laura Mechtouff
- Stroke Department, Hospices Civils de Lyon, France; CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - René Buchet
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Eva Hamade
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| | - Caroline Fonta
- Brain and Cognition Research Center CerCo, CNRS UMR5549, Université de Toulouse, France
| | - Emmanuelle Canet-Soulas
- CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | | | - David Magne
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France.
| |
Collapse
|
6
|
Kahn C, Singh S, Mathew R, Ramrattan LA, Mohammed IJ, Omman R. Rituximab Used for the Treatment of Nonuremic Calciphylaxis: A Complication of Prolonged Steroid Use in Lupus Nephritis. Cureus 2022; 14:e26516. [PMID: 35795578 PMCID: PMC9250422 DOI: 10.7759/cureus.26516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2022] [Indexed: 11/05/2022] Open
Abstract
Nonuremic calciphylaxis (NUC) is a rare and debilitating form of panniculitis. NUC is associated with a high mortality rate within the first year of diagnosis. Connective tissue diseases account for a small fraction of the reported cases. However, there have also been reported cases of patients developing NUC while on treatment with chronic corticosteroid immunosuppressive therapy. The pathophysiology of NUC is still not fully established. Several risk factors including underlying diseases, obesity, female gender, and medications have been associated with the development of NUC. The diagnosis remains challenging due to the condition's similarities with other forms of panniculitis. The gold standard for diagnosis is a tissue biopsy showing calcifications within the medial layer of arterioles and the presence of microthrombi with surrounding necrosis. The treatment for NUC has not advanced much in recent years and focuses on the management of the underlying condition, wound care, and treating any superimposed infection. Treating superimposed infections remains important as most of the associated mortality from NUC occurs due to sepsis. We describe a case of a young woman with lupus nephritis who developed NUC while on prolonged corticosteroid therapy. She did not respond to several immunosuppressive agents and was ultimately treated with rituximab, a monoclonal antibody against CD20 antigen, as salvage therapy.
Collapse
|
7
|
Zhang M, Li T, Tu Z, Zhang Y, Wang X, Zang D, Xu D, Feng Y, He F, Ni M, Wang D, Zhou H. Both high glucose and phosphate overload promote senescence-associated calcification of vascular muscle cells. Int Urol Nephrol 2022; 54:2719-2731. [PMID: 35396645 DOI: 10.1007/s11255-022-03195-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 01/11/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE The NAD+-dependent deacetylase, sirtuin 1 (SIRT1), plays an important role in vascular calcification induced by high glucose and/or high phosphate levels. However, the mechanism by which SIRT1 regulates this process is still not fully understood. Thus, this study aimed to determine the role of high glucose and phosphate in vascular calcification and the molecular mechanisms underlying SIRT1 regulation. METHODS Vascular smooth muscle cells (VSMCs) were cultured under normal, high phosphate, and/or high-glucose conditions for 9 days. Alizarin red staining and calcification content analyses were used to determine calcium deposition. VSMC senescence was detected by β-galactosidase (SA-β-Gal) staining and p21 expression. RESULTS Mouse VSMCs exposed to high phosphate and high glucose in vitro showed increased calcification, which was correlated with the induction of cell senescence, as confirmed by the increased SA-β-galactosidase activity and p21 expression. SRT1720, an activator of SIRT1, inhibits p65 acetylation, the nuclear factor-κ-gene binding (NF-κB) pathway, and VSMC transdifferentiation, prevents senescence and reactive oxygen species (ROS) production, and reduces vascular calcification. In contrast, sirtinol, an inhibitor of SIRT1, increases p65 acetylation, activates the NF-κB pathway, induces vascular smooth muscle cell transdifferentiation and senescence, and promotes vascular calcification. CONCLUSIONS High glucose and high phosphate levels induce senescence and vascular calcification in VSMCs, and the combined effect of high glucose and phosphate can inhibit SIRT1 expression. SIRT1 inhibits vascular smooth muscle cell senescence and osteogenic differentiation by inhibiting NF-κB activity, thereby inhibiting vascular calcification.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tianyu Li
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhenzhen Tu
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China
| | - Yuying Zhang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China
| | - Xuerong Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dandan Zang
- The Center for Scientific Research of Anhui Medical University, Hefei, China
| | - Deping Xu
- Department of Clinical Laboratory, Affiliated Hefei Hospital of Anhui Medical University, Hefei, China
| | - Yang Feng
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China
| | - Fan He
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China
| | - Mingyue Ni
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China
| | - Deguang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Haisheng Zhou
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China.,The Center for Scientific Research of Anhui Medical University, Hefei, China
| |
Collapse
|
8
|
Sun XJ, Liu NF. Diabetic mellitus, vascular calcification and hypoxia: A complex and neglected tripartite relationship. Cell Signal 2021; 91:110219. [PMID: 34921978 DOI: 10.1016/j.cellsig.2021.110219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/11/2021] [Accepted: 12/11/2021] [Indexed: 11/15/2022]
Abstract
DM (diabetic mellitus) and its common vascular complications VC (vascular calcification), are increasingly harmful to human health. In recent years, the research on the relationship between DM and VC is also deepening. Hypoxia, as one of the pathogenic factors of many disease models, is also closely related to the occurrence of DM and VC. There are some studies on the role of hypoxia in the pathogenesis of DM and VC respectively, but no one has made an in-depth summary of the systematic connection between hypoxia, DM and VC. Therefore, what we want to review in this article are the relationship between DM, VC and hypoxia, respectively, as well as the role of hypoxia in the development of DM and VC, which has little concern but is a novel and potentially target that may provide some new ideas for the prevention and treatment of DM, VC, especially diabetic VC.
Collapse
Affiliation(s)
- Xue-Jiao Sun
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing 210009, PR China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing 210009, PR China.
| |
Collapse
|
9
|
Henze LA, Estepa M, Pieske B, Lang F, Eckardt KU, Alesutan I, Voelkl J. Zinc Ameliorates the Osteogenic Effects of High Glucose in Vascular Smooth Muscle Cells. Cells 2021; 10:cells10113083. [PMID: 34831306 PMCID: PMC8623153 DOI: 10.3390/cells10113083] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/23/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
In diabetic patients, medial vascular calcification is common and associated with increased cardiovascular mortality. Excessive glucose concentrations can activate the nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-kB) and trigger pro-calcific effects in vascular smooth muscle cells (VSMCs), which may actively augment vascular calcification. Zinc is able to mitigate phosphate-induced VSMC calcification. Reduced serum zinc levels have been reported in diabetes mellitus. Therefore, in this study the effects of zinc supplementation were investigated in primary human aortic VSMCs exposed to excessive glucose concentrations. Zinc treatment was found to abrogate the stimulating effects of high glucose on VSMC calcification. Furthermore, zinc was found to blunt the increased expression of osteogenic and chondrogenic markers in high glucose-treated VSMCs. High glucose exposure was shown to activate NF-kB in VSMCs, an effect that was blunted by additional zinc treatment. Zinc was further found to increase the expression of TNFα-induced protein 3 (TNFAIP3) in high glucose-treated VSMCs. The silencing of TNFAIP3 was shown to abolish the protective effects of zinc on high glucose-induced NF-kB-dependent transcriptional activation, osteogenic marker expression, and the calcification of VSMCs. Silencing of the zinc-sensing receptor G protein-coupled receptor 39 (GPR39) was shown to abolish zinc-induced TNFAIP3 expression and the effects of zinc on high glucose-induced osteogenic marker expression. These observations indicate that zinc may be a protective factor during vascular calcification in hyperglycemic conditions.
Collapse
Affiliation(s)
- Laura A. Henze
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany; (L.A.H.); (M.E.); (B.P.)
| | - Misael Estepa
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany; (L.A.H.); (M.E.); (B.P.)
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany; (L.A.H.); (M.E.); (B.P.)
| | - Florian Lang
- Department of Vegetative and Clinical Physiology, Eberhard Karls University Tübingen, 72076 Tübingen, Germany;
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.-U.E.); (J.V.)
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, 4040 Linz, Austria
- Correspondence: ; Tel.: +43-732-2468-8990
| | - Jakob Voelkl
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.-U.E.); (J.V.)
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, 4040 Linz, Austria
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347 Berlin, Germany
| |
Collapse
|
10
|
Samara VA, Das S, Reddy MA, Tanwar VS, Stapleton K, Leung A, Abdollahi M, Ganguly R, Lanting L, Natarajan R. Angiotensin II-Induced Long Non-Coding RNA Alivec Regulates Chondrogenesis in Vascular Smooth Muscle Cells. Cells 2021; 10:2696. [PMID: 34685676 PMCID: PMC8535098 DOI: 10.3390/cells10102696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) play key roles in Angiotensin II (AngII) signaling but their role in chondrogenic transformation of vascular smooth muscle cells (VSMCs) is unknown. We describe a novel AngII-induced lncRNA Alivec (Angiotensin II-induced lncRNA in VSMCs eliciting chondrogenic phenotype) implicated in VSMC chondrogenesis. In rat VSMCs, Alivec and the nearby gene Acan, a chondrogenic marker, were induced by growth factors AngII and PDGF and the inflammatory cytokine TNF-α. AngII co-regulated Alivec and Acan through the activation of AngII type1 receptor signaling and Sox9, a master transcriptional regulator of chondrogenesis. Alivec knockdown with GapmeR antisense-oligonucleotides attenuated the expression of AngII-induced chondrogenic marker genes, including Acan, and inhibited the chondrogenic phenotype of VSMCs. Conversely, Alivec overexpression upregulated these genes and promoted chondrogenic transformation. RNA-pulldown coupled to mass-spectrometry identified Tropomyosin-3-alpha and hnRNPA2B1 proteins as Alivec-binding proteins in VSMCs. Furthermore, male rats with AngII-driven hypertension showed increased aortic expression of Alivec and Acan. A putative human ortholog ALIVEC, was induced by AngII in human VSMCs, and this locus was found to harbor the quantitative trait loci affecting blood pressure. Together, these findings suggest that AngII-regulated lncRNA Alivec functions, at least in part, to mediate the AngII-induced chondrogenic transformation of VSMCs implicated in vascular dysfunction and hypertension.
Collapse
MESH Headings
- Aggrecans/genetics
- Aggrecans/metabolism
- Angiotensin II/pharmacology
- Animals
- Aorta/metabolism
- Blood Pressure/drug effects
- Blood Pressure/genetics
- Chondrogenesis/drug effects
- Chondrogenesis/genetics
- Enhancer Elements, Genetic/genetics
- Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism
- Humans
- Male
- Muscle Contraction/genetics
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Osteogenesis/drug effects
- Osteogenesis/genetics
- Phenotype
- Quantitative Trait Loci/genetics
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- SOX9 Transcription Factor/metabolism
- Tropomyosin/metabolism
- Up-Regulation/drug effects
- Up-Regulation/genetics
- src-Family Kinases/metabolism
- Rats
Collapse
Affiliation(s)
- Vishnu Amaram Samara
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA 91010, USA; (V.A.S.); (S.D.); (M.A.R.); (V.S.T.); (K.S.); (A.L.); (M.A.); (R.G.); (L.L.)
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Sadhan Das
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA 91010, USA; (V.A.S.); (S.D.); (M.A.R.); (V.S.T.); (K.S.); (A.L.); (M.A.); (R.G.); (L.L.)
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, UP 226031, India
| | - Marpadga A. Reddy
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA 91010, USA; (V.A.S.); (S.D.); (M.A.R.); (V.S.T.); (K.S.); (A.L.); (M.A.); (R.G.); (L.L.)
| | - Vinay Singh Tanwar
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA 91010, USA; (V.A.S.); (S.D.); (M.A.R.); (V.S.T.); (K.S.); (A.L.); (M.A.); (R.G.); (L.L.)
| | - Kenneth Stapleton
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA 91010, USA; (V.A.S.); (S.D.); (M.A.R.); (V.S.T.); (K.S.); (A.L.); (M.A.); (R.G.); (L.L.)
| | - Amy Leung
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA 91010, USA; (V.A.S.); (S.D.); (M.A.R.); (V.S.T.); (K.S.); (A.L.); (M.A.); (R.G.); (L.L.)
| | - Maryam Abdollahi
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA 91010, USA; (V.A.S.); (S.D.); (M.A.R.); (V.S.T.); (K.S.); (A.L.); (M.A.); (R.G.); (L.L.)
| | - Rituparna Ganguly
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA 91010, USA; (V.A.S.); (S.D.); (M.A.R.); (V.S.T.); (K.S.); (A.L.); (M.A.); (R.G.); (L.L.)
| | - Linda Lanting
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA 91010, USA; (V.A.S.); (S.D.); (M.A.R.); (V.S.T.); (K.S.); (A.L.); (M.A.); (R.G.); (L.L.)
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Duarte, CA 91010, USA; (V.A.S.); (S.D.); (M.A.R.); (V.S.T.); (K.S.); (A.L.); (M.A.); (R.G.); (L.L.)
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
11
|
Osteoclast-Mediated Cell Therapy as an Attempt to Treat Elastin Specific Vascular Calcification. Molecules 2021; 26:molecules26123643. [PMID: 34203711 PMCID: PMC8232296 DOI: 10.3390/molecules26123643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 01/03/2023] Open
Abstract
Inflammation and stiffness in the arteries is referred to as vascular calcification. This process is a prevalent yet poorly understood consequence of cardiovascular disease and diabetes mellitus, comorbidities with few treatments clinically available. Because this is an active process similar to bone formation, it is hypothesized that osteoclasts (OCs), bone-resorbing cells in the body, could potentially work to reverse existing calcification by resorbing bone material. The receptor activator of nuclear kappa B-ligand (RANKL) is a molecule responsible for triggering a response in monocytes and macrophages that allows them to differentiate into functional OCs. In this study, OC and RANKL delivery were employed to determine whether calcification could be attenuated. OCs were either delivered via direct injection, collagen/alginate microbeads, or collagen gel application, while RANKL was delivered via injection, through either a porcine subdermal model or aortic injury model. While in vitro results yielded a decrease in calcification using OC therapy, in vivo delivery mechanisms did not provide control or regulation to keep cells localized long enough to induce calcification reduction. However, these results do provide context and direction for the future of OC therapy, revealing necessary steps for this treatment to effectively reduce calcification in vivo. The discrepancy between in vivo and in vitro success for OC therapy points to the need for a more stable and time-controlled delivery mechanism that will allow OCs not only to remain at the site of calcification, but also to be regulated so that they are healthy and functioning normally when introduced to diseased tissue.
Collapse
|
12
|
DeClue C, Chinnakotla B, Gardner MJ. Non-Uremic Calciphylaxis: An Unexpected Complication With Recombinant Human Parathyroid Hormone. Cureus 2021; 13:e15014. [PMID: 34150377 PMCID: PMC8202451 DOI: 10.7759/cureus.15014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Calciphylaxis is a rare syndrome of calcific microvascular occlusion, whereas non-uremic calciphylaxis (NUC) is a subset of this disease in which renal impairment is not observed. Recombinant human parathyroid hormone (rhPTH) (1-84) is a medication approved for the management of hypocalcemia in patients with hypoparathyroidism. We present a case report of a 38-year-old woman with postoperative hypoparathyroidism treated with rhPTH who subsequently developed calciphylactic lesions on her abdomen. Multidisciplinary interventions included intravenous and intralesional sodium thiosulfate therapy, laboratory monitoring, dermatological wound care, and pain management. Calciphylaxis can rarely be precipitated by rhPTH due to its effect on calcium and phosphorus balance even in the setting of normal renal function. The use of calcium and calcitriol supplementation, complicated by factors such as female sex and obesity, may have contributed in this patient’s case. Hence, regular follow-up with tapering off of calcium and calcitriol supplementation is important in patients receiving rhPTH.
Collapse
Affiliation(s)
- Cory DeClue
- Internal Medicine, University of Missouri, Columbia, USA
| | | | | |
Collapse
|
13
|
Phadwal K, Vrahnas C, Ganley IG, MacRae VE. Mitochondrial Dysfunction: Cause or Consequence of Vascular Calcification? Front Cell Dev Biol 2021; 9:611922. [PMID: 33816463 PMCID: PMC8010668 DOI: 10.3389/fcell.2021.611922] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/04/2021] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are crucial bioenergetics powerhouses and biosynthetic hubs within cells, which can generate and sequester toxic reactive oxygen species (ROS) in response to oxidative stress. Oxidative stress-stimulated ROS production results in ATP depletion and the opening of mitochondrial permeability transition pores, leading to mitochondria dysfunction and cellular apoptosis. Mitochondrial loss of function is also a key driver in the acquisition of a senescence-associated secretory phenotype that drives senescent cells into a pro-inflammatory state. Maintaining mitochondrial homeostasis is crucial for retaining the contractile phenotype of the vascular smooth muscle cells (VSMCs), the most prominent cells of the vasculature. Loss of this contractile phenotype is associated with the loss of mitochondrial function and a metabolic shift to glycolysis. Emerging evidence suggests that mitochondrial dysfunction may play a direct role in vascular calcification and the underlying pathologies including (1) impairment of mitochondrial function by mineral dysregulation i.e., calcium and phosphate overload in patients with end-stage renal disease and (2) presence of increased ROS in patients with calcific aortic valve disease, atherosclerosis, type-II diabetes and chronic kidney disease. In this review, we discuss the cause and consequence of mitochondrial dysfunction in vascular calcification and underlying pathologies; the role of autophagy and mitophagy pathways in preventing mitochondrial dysfunction during vascular calcification and finally we discuss mitochondrial ROS, DRP1, and HIF-1 as potential novel markers and therapeutic targets for maintaining mitochondrial homeostasis in vascular calcification.
Collapse
Affiliation(s)
- Kanchan Phadwal
- Functional Genetics and Development Division, The Roslin Institute and The Royal (Dick) School of Veterinary Studies (R(D)SVS), University of Edinburgh, Midlothian, United Kingdom
| | - Christina Vrahnas
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, University of Dundee, Dundee, United Kingdom
| | - Ian G. Ganley
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, University of Dundee, Dundee, United Kingdom
| | - Vicky E. MacRae
- Functional Genetics and Development Division, The Roslin Institute and The Royal (Dick) School of Veterinary Studies (R(D)SVS), University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
14
|
Buchet R, Tribes C, Rouaix V, Doumèche B, Fiore M, Wu Y, Magne D, Mebarek S. Hydrolysis of Extracellular ATP by Vascular Smooth Muscle Cells Transdifferentiated into Chondrocytes Generates P i but Not PP i. Int J Mol Sci 2021; 22:ijms22062948. [PMID: 33799449 PMCID: PMC8000465 DOI: 10.3390/ijms22062948] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Tissue non-specific alkaline phosphatase (TNAP) is suspected to induce atherosclerosis plaque calcification. TNAP, during physiological mineralization, hydrolyzes the mineralization inhibitor inorganic pyrophosphate (PPi). Since atherosclerosis plaques are characterized by the presence of necrotic cells that probably release supraphysiological concentrations of ATP, we explored whether this extracellular adenosine triphosphate (ATP) is hydrolyzed into the mineralization inhibitor PPi or the mineralization stimulator inorganic phosphate (Pi), and whether TNAP is involved. (2) Methods: Murine aortic smooth muscle cell line (MOVAS cells) were transdifferentiated into chondrocyte-like cells in calcifying medium, containing ascorbic acid and β-glycerophosphate. ATP hydrolysis rates were determined in extracellular medium extracted from MOVAS cultures during their transdifferentiation, using 31P-NMR and IR spectroscopy. (3) Results: ATP and PPi hydrolysis by MOVAS cells increased during transdifferentiation. ATP hydrolysis was sequential, yielding adenosine diphosphate (ADP), adenosine monophosphate (AMP), and adenosine without any detectable PPi. The addition of levamisole partially inhibited ATP hydrolysis, indicating that TNAP and other types of ectonucleoside triphoshatediphosphohydrolases contributed to ATP hydrolysis. (4) Conclusions: Our findings suggest that high ATP levels released by cells in proximity to vascular smooth muscle cells (VSMCs) in atherosclerosis plaques generate Pi and not PPi, which may exacerbate plaque calcification.
Collapse
Affiliation(s)
- Rene Buchet
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, Université Lyon 1, French National Centre for Scientific Research, F-69622 Lyon, France; (C.T.); (V.R.); (B.D.); (M.F.); (D.M.); (S.M.)
- Correspondence:
| | - Camille Tribes
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, Université Lyon 1, French National Centre for Scientific Research, F-69622 Lyon, France; (C.T.); (V.R.); (B.D.); (M.F.); (D.M.); (S.M.)
| | - Valentine Rouaix
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, Université Lyon 1, French National Centre for Scientific Research, F-69622 Lyon, France; (C.T.); (V.R.); (B.D.); (M.F.); (D.M.); (S.M.)
| | - Bastien Doumèche
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, Université Lyon 1, French National Centre for Scientific Research, F-69622 Lyon, France; (C.T.); (V.R.); (B.D.); (M.F.); (D.M.); (S.M.)
| | - Michele Fiore
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, Université Lyon 1, French National Centre for Scientific Research, F-69622 Lyon, France; (C.T.); (V.R.); (B.D.); (M.F.); (D.M.); (S.M.)
| | - Yuqing Wu
- State Key Laboratory of Supramolecular Structure and Materials, Institute of Theoretical Chemistry, Jilin University, Changchun 130012, China;
| | - David Magne
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, Université Lyon 1, French National Centre for Scientific Research, F-69622 Lyon, France; (C.T.); (V.R.); (B.D.); (M.F.); (D.M.); (S.M.)
| | - Saida Mebarek
- Institute for Molecular and Supramolecular Chemistry and Biochemistry, Université Lyon 1, French National Centre for Scientific Research, F-69622 Lyon, France; (C.T.); (V.R.); (B.D.); (M.F.); (D.M.); (S.M.)
| |
Collapse
|
15
|
Reinhold S, Blankesteijn WM, Foulquier S. The Interplay of WNT and PPARγ Signaling in Vascular Calcification. Cells 2020; 9:cells9122658. [PMID: 33322009 PMCID: PMC7763279 DOI: 10.3390/cells9122658] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/02/2022] Open
Abstract
Vascular calcification (VC), the ectopic deposition of calcium phosphate crystals in the vessel wall, is one of the primary contributors to cardiovascular death. The pathology of VC is determined by vascular topography, pre-existing diseases, and our genetic heritage. VC evolves from inflammation, mediated by macrophages, and from the osteochondrogenic transition of vascular smooth muscle cells (VSMC) in the atherosclerotic plaque. This pathologic transition partly resembles endochondral ossification, involving the chronologically ordered activation of the β-catenin-independent and -dependent Wingless and Int-1 (WNT) pathways and the termination of peroxisome proliferator-activated receptor γ (PPARγ) signal transduction. Several atherosclerotic plaque studies confirmed the differential activity of PPARγ and the WNT signaling pathways in VC. Notably, the actively regulated β-catenin-dependent and -independent WNT signals increase the osteochondrogenic transformation of VSMC through the up-regulation of the osteochondrogenic transcription factors SRY-box transcription factor 9 (SOX9) and runt-related transcription factor 2 (RUNX2). In addition, we have reported studies showing that WNT signaling pathways may be antagonized by PPARγ activation via the expression of different families of WNT inhibitors and through its direct interaction with β-catenin. In this review, we summarize the existing knowledge on WNT and PPARγ signaling and their interplay during the osteochondrogenic differentiation of VSMC in VC. Finally, we discuss knowledge gaps on this interplay and its possible clinical impact.
Collapse
Affiliation(s)
- Stefan Reinhold
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (S.R.); (W.M.B.)
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (S.R.); (W.M.B.)
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (S.R.); (W.M.B.)
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-433881409
| |
Collapse
|
16
|
Rapp N, Evenepoel P, Stenvinkel P, Schurgers L. Uremic Toxins and Vascular Calcification-Missing the Forest for All the Trees. Toxins (Basel) 2020; 12:E624. [PMID: 33003628 PMCID: PMC7599869 DOI: 10.3390/toxins12100624] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 12/23/2022] Open
Abstract
The cardiorenal syndrome relates to the detrimental interplay between the vascular system and the kidney. The uremic milieu induced by reduced kidney function alters the phenotype of vascular smooth muscle cells (VSMC) and promotes vascular calcification, a condition which is strongly linked to cardiovascular morbidity and mortality. Biological mechanisms involved include generation of reactive oxygen species, inflammation and accelerated senescence. A better understanding of the vasotoxic effects of uremic retention molecules may reveal novel avenues to reduce vascular calcification in CKD. The present review aims to present a state of the art on the role of uremic toxins in pathogenesis of vascular calcification. Evidence, so far, is fragmentary and limited with only a few uremic toxins being investigated, often by a single group of investigators. Experimental heterogeneity furthermore hampers comparison. There is a clear need for a concerted action harmonizing and standardizing experimental protocols and combining efforts of basic and clinical researchers to solve the complex puzzle of uremic vascular calcification.
Collapse
MESH Headings
- Animals
- Cardio-Renal Syndrome/metabolism
- Cardio-Renal Syndrome/pathology
- Cardio-Renal Syndrome/physiopathology
- Cardio-Renal Syndrome/therapy
- Humans
- Kidney/metabolism
- Kidney/pathology
- Kidney/physiopathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Prognosis
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/physiopathology
- Renal Insufficiency, Chronic/therapy
- Toxins, Biological/metabolism
- Uremia/metabolism
- Uremia/pathology
- Uremia/physiopathology
- Uremia/therapy
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/physiopathology
- Vascular Calcification/therapy
Collapse
Affiliation(s)
- Nikolas Rapp
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Pieter Evenepoel
- Laboratory of Nephrology, KU Leuven Department of Microbiology and Immunology, University Hospitals Leuven, 3000 Leuven, Belgium;
| | - Peter Stenvinkel
- Karolinska Institute, Department of Clinical Science, Intervention and Technology, Division of Renal Medicine, 141 86 Stockholm, Sweden;
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
17
|
Role of SGK1 in the Osteogenic Transdifferentiation and Calcification of Vascular Smooth Muscle Cells Promoted by Hyperglycemic Conditions. Int J Mol Sci 2020; 21:ijms21197207. [PMID: 33003561 PMCID: PMC7583813 DOI: 10.3390/ijms21197207] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
In diabetes mellitus, hyperglycemia promotes the osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs) to enhance medial vascular calcification, a common complication strongly associated with cardiovascular disease and mortality. The mechanisms involved are, however, still poorly understood. Therefore, the present study explored the potential role of serum- and glucocorticoid-inducible kinase 1 (SGK1) during vascular calcification promoted by hyperglycemic conditions. Exposure to high-glucose conditions up-regulated the SGK1 expression in primary human aortic VSMCs. High glucose increased osteogenic marker expression and activity and, thus, promoted the osteogenic transdifferentiation of VSMCs, effects significantly suppressed by additional treatment with the SGK1 inhibitor EMD638683. Moreover, high glucose augmented the mineralization of VSMCs in the presence of calcification medium, effects again significantly reduced by SGK1 inhibition. Similarly, SGK1 knockdown blunted the high glucose-induced osteogenic transdifferentiation of VSMCs. The osteoinductive signaling promoted by high glucose required SGK1-dependent NF-kB activation. In addition, advanced glycation end products (AGEs) increased the SGK1 expression in VSMCs, and SGK1 inhibition was able to interfere with AGEs-induced osteogenic signaling. In conclusion, SGK1 is up-regulated and mediates, at least partly, the osteogenic transdifferentiation and calcification of VSMCs during hyperglycemic conditions. Thus, SGK1 inhibition may reduce the development of vascular calcification promoted by hyperglycemia in diabetes.
Collapse
|
18
|
Han L, Zhang Y, Zhang M, Guo L, Wang J, Zeng F, Xu D, Yin Z, Xu Y, Wang D, Zhou H. Interleukin-1β-Induced Senescence Promotes Osteoblastic Transition of Vascular Smooth Muscle Cells. Kidney Blood Press Res 2020; 45:314-330. [PMID: 32126555 DOI: 10.1159/000504298] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 10/21/2019] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Interleukin (IL)-1β, as a key biomarker and mediator of vascular calcification in patients with end-stage renal disease (ESRD), may be involved in the process of premature senescence of vascular smooth muscle cells (VSMCs). This work sought to investigate whether IL-1β-induced premature senescence contributes to the process of osteoblastic transition and vascular calcification in VSMCs. METHODS Eighty-eight patients with ESRD (aged 25-81 years), 11 healthy individuals, and 15 cases of lesion-free distal radial arteries from dialysis ESRD patients with angiostomy were collected in this study. Immunohistochemical analysis was performed to detect expression of IL-1β, p21, and bone morphogenetic protein-2 (BMP2) in the distal radial arteries. Primary human VSMCs from healthy neonatal umbilical cords were incubated with test agents for 1-3 days. Intracellular levels of reactive oxygen species (ROS) and senescence-associated-β-galactosidase (SA-β-gal) staining were used to detect senescent cells. Alizarin red staining and the calcium content of the cell layer were used to detect mineral deposition in VSMCs. RESULTS Coincident with positive staining of IL-1β, p21, and BMP2 in the lesion-free distal radial arteries, 66.67% patients showed mineral deposition. Serum IL-1β was 0.24 ± 0.57, 1.20 ± 2.95, and 9.41 ± 40.52 pg/mL in 11 healthy individuals, 20 patients without calcification, and 53 patients with calcification, respectively. Analysis of the cross-table chi-square test showed cardiovascular calcification is not correlated with levels of serum IL-1β in patients with ESRD (p = 0.533). In response to IL-1β, VSMCs showed a senescence-like phenotype, such as flat and enlarged morphology, increased expression of p21, an increased activity of SA-β-gal, and increased levels of ROS. IL-1β-induced senescence of VSMCs was required for the activation of IL-1β/NF-κB/p53/p21 signaling pathway. IL-1β-induced senescent VSMCs underwent calcification due to osteoblastic transition mainly depending upon the upregulation of BMP2. Resveratrol, an activator of sirtuin-1, postponed the IL-1β-induced senescence through blocking the NF-κB/p53/p21 pathway and attenuated the osteoblastic transition and calcification in VSMCs. CONCLUSIONS High levels of IL-1β in medial smooth muscles of arteries may play roles in inducing senescence-associated calcification. IL-1β-induced senescence depending on the activation of the NF-κB/p53/p21 signaling pathway and contributing to osteoblastic transition of VSMCs.
Collapse
Affiliation(s)
- Linzi Han
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China.,Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuying Zhang
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China.,Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingming Zhang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liyu Guo
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China
| | - Jun Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fanjun Zeng
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China
| | - Deping Xu
- Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zongzhi Yin
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanhong Xu
- Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Deguang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China,
| | - Haisheng Zhou
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, China.,The Center for Scientific Research of Anhui Medical University, Hefei, China
| |
Collapse
|
19
|
Ding X, Chen J, Wu C, Wang G, Zhou C, Chen S, Wang K, Zhang A, Ye P, Wu J, Chen S, Zhang H, Xu K, Wang S, Xia J. Nucleotide-Binding Oligomerization Domain-Like Receptor Protein 3 Deficiency in Vascular Smooth Muscle Cells Prevents Arteriovenous Fistula Failure Despite Chronic Kidney Disease. J Am Heart Assoc 2020; 8:e011211. [PMID: 30587058 PMCID: PMC6405733 DOI: 10.1161/jaha.118.011211] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background The arteriovenous fistula (AVF) is the preferred hemodialysis access for patients with chronic kidney disease. Chronic kidney disease can increase neointima formation, which greatly contributes to AVF failure by an unknown mechanism. Our study aimed to determine the role of nucleotide‐binding oligomerization domain‐like receptor protein 3 (NLRP3) in neointima formation induced by experimental AVFs in the presence of chronic kidney disease. Methods and Results From our findings, NLRP3 was upregulated in the intimal lesions of AVFs in both uremic mice and patients. Smooth muscle–specific knockout NLRP3 mice exhibited markedly decreased neointima formation in the outflow vein of AVFs. Compared with primary vascular smooth muscle cells isolated from control mice, those isolated from smooth muscle–specific knockout NLRP3 mice showed compromised proliferation, migration, phenotypic switching, and a weakened ability to activate mononuclear macrophages. To identify how NLRP3 functions, several small‐molecule inhibitors were used. The results showed that NLRP3 regulates smooth muscle cell proliferation and migration through Smad2/3 phosphorylation rather than through caspase‐1/interleukin‐1 signaling. Unexpectedly, the selective NLRP3‐inflammasome inhibitor MCC950 also repressed Smad2/3 phosphorylation and relieved chronic kidney disease–promoted AVF failure independent of macrophages. Conclusions Our findings suggest that NLRP3 in vascular smooth muscle cells may play a crucial role in uremia‐associated AVF failure and may be a promising therapeutic target for the treatment of AVF failure.
Collapse
Affiliation(s)
- Xiangchao Ding
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jiuling Chen
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Chuangyan Wu
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China.,2 Department of Thoracic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Guohua Wang
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Cheng Zhou
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Shanshan Chen
- 3 Key Laboratory for Molecular Diagnosis of Hubei Province Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China.,4 Central Laboratory Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ke Wang
- 6 Department of Respiratory and Critical Care Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Anchen Zhang
- 5 Department of Cardiovascular Medicine Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ping Ye
- 5 Department of Cardiovascular Medicine Central Hospital of Wuhan Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jie Wu
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Shanshan Chen
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Hao Zhang
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Kaiying Xu
- 2 Department of Thoracic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Sihua Wang
- 2 Department of Thoracic Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jiahong Xia
- 1 Department of Cardiovascular Surgery Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
20
|
Chang Z, Yan G, Yan H, Zheng J, Liu Z. Reticulocalbin 2 enhances osteogenic differentiation of human vascular smooth muscle cells in diabetic conditions. Life Sci 2019; 233:116746. [DOI: 10.1016/j.lfs.2019.116746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/19/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022]
|
21
|
Zhou YB, Zhou H, Li L, Kang Y, Cao X, Wu ZY, Ding L, Sethi G, Bian JS. Hydrogen Sulfide Prevents Elastin Loss and Attenuates Calcification Induced by High Glucose in Smooth Muscle Cells through Suppression of Stat3/Cathepsin S Signaling Pathway. Int J Mol Sci 2019; 20:ijms20174202. [PMID: 31461977 PMCID: PMC6747320 DOI: 10.3390/ijms20174202] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/12/2019] [Accepted: 08/19/2019] [Indexed: 02/06/2023] Open
Abstract
Vascular calcification can be enhanced by hyperglycemia. Elastin loss in tunica media promotes the osteogenic transformation of smooth muscle cells (SMCs) and involves arterial medial calcification (AMC) that is associated with a high incidence of cardiovascular risk in patients with type 2 diabetes. Here, we tested whether hydrogen sulfide (H2S), an endogenous gaseous mediator, can prevent elastin loss and attenuate calcification induced by high glucose in SMCs. Calcification was induced by high glucose (4500 mg/L) in human aortic SMCs (HASMCs) under the condition of calcifying medium containing 10 mM β-glycerophosphate (β-GP). The experiments showed that NaHS (an H2S donor, 100 μM) mitigated the calcification of HASMCs treated with high glucose by decreasing calcium and phosphorus levels, calcium deposition and ALP activity and inhibited osteogenic transformation by increasing SMα-actin and SM22α, two phenotypic markers of smooth muscle cells, and decreasing core binding factor α-1 (Cbfα-1), a key factor in bone formation, protein expressions in HASMCs. Moreover, NaHS administration inhibited the activation of Stat3, cathepsin S (CAS) activity and its expression, but increased the level of elastin protein. Pharmacological inhibition or gene silencing Stat3 not only reversed elastin loss, but also attenuated CAS expression. Inhibition of CAS alleviated, while CAS overexpression exacerbated, elastin loss. Interestingly, overexpression of wild type (WT)-Stat3, but not its mutant C259S, elevated CAS protein expression and reduced elastin level. Moreover, NaHS induced S-sulfhydration in WT, but not in the C259S Stat3. These data suggest that H2S may directly regulate Cys259 residue in Stat3 and then impair its signaling function. Our data indicate that H2S may attenuate vascular calcification by upregulating elastin level through the inhibition of Stat3/CAS signaling.
Collapse
Affiliation(s)
- Ye-Bo Zhou
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Hong Zhou
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Li Li
- Department of Pathophysiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Ying Kang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Xu Cao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Zhi-Yuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Lei Ding
- Department of Pathophysiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
- National University of Singapore (Suzhou) Research Institute (NUSRI), Suzhou Industrial Park, Suzhou 215123, China.
| |
Collapse
|
22
|
Castelblanco M, Nasi S, Pasch A, So A, Busso N. The role of the gasotransmitter hydrogen sulfide in pathological calcification. Br J Pharmacol 2019; 177:778-792. [PMID: 31231793 DOI: 10.1111/bph.14772] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022] Open
Abstract
Calcification is the deposition of minerals, mainly hydroxyapatite, inside the cell or in the extracellular matrix. Physiological calcification is central for many aspects of development including skeletal and tooth growth; conversely, pathological mineralization occurs in soft tissues and is significantly associated with malfunction and impairment of the tissue where it is located. Various mechanisms have been proposed to explain calcification. However, this research area lacks a more integrative, systemic, and global perspective that could explain both physiological and pathological processes. In this review, we propose such an integrated explanation. Hydrogen sulfide (H2 S) is a newly recognized multifunctional gasotransmitters and tis actions have been studied in different physiological and pathological contexts, but little is known about its potential role on calcification. Interestingly, we found that H2 S promotes calcification under physiological conditions and has an inhibitory effect on pathological processes. This makes H2 S a potential therapy for diseases related to pathological calcification. LINKED ARTICLES: This article is part of a themed section on Hydrogen Sulfide in Biology & Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.4/issuetoc.
Collapse
Affiliation(s)
- Mariela Castelblanco
- Service of Rheumatology, DAL, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Sonia Nasi
- Service of Rheumatology, DAL, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| | | | - Alexander So
- Service of Rheumatology, DAL, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Nathalie Busso
- Service of Rheumatology, DAL, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
23
|
Bochkareva EV, Kim IV, Butina EK, Stulin ID, Trukhanov SA, Rudenko BA, Boytsov SA, Drapkina OM. Mammographic Screening as a Tool for Cardiovascular Risk Assessing. Part 1. Breast Arterial Calcification: Pathomorphology, Prevalence and Risk Factors. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2019. [DOI: 10.20996/1819-6446-2019-15-2-244-250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
| | - I. V. Kim
- National Medical Research Center for Preventive Medicine
| | - E. K. Butina
- National Medical Research Center for Preventive Medicine
| | - I. D. Stulin
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry
| | - S. A. Trukhanov
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry
| | - B. A. Rudenko
- National Medical Research Center for Preventive Medicine
| | | | - O. M. Drapkina
- National Medical Research Center for Preventive Medicine
| |
Collapse
|
24
|
Barchetta I, Ceccarelli V, Cimini FA, Bertoccini L, Fraioli A, Alessandri C, Lenzi A, Baroni MG, Cavallo MG. Impaired bone matrix glycoprotein pattern is associated with increased cardio-metabolic risk profile in patients with type 2 diabetes mellitus. J Endocrinol Invest 2019; 42:513-520. [PMID: 30132286 DOI: 10.1007/s40618-018-0941-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE Osteopontin (OPN), osteoprotegerin (OPG) and osteocalcin (OC) are matrix glycoproteins which mediate bone mineralization; moreover, their effects on glucose/insulin homeostasis have recently been demonstrated. Higher circulating OPN and OPG levels have been associated with the presence of insulin resistance, atherosclerosis and coronary heart disease. No data are available on contextual changes of these markers in type 2 diabetes mellitus (T2DM). Therefore, aims of this study were to evaluate serum OPN, OPG and OC levels in T2DM patients and their clinical correlates. METHODS We recruited 83 consecutive T2DM patients referring to our diabetes outpatient clinics at Sapienza, University of Rome, and 71 non-diabetic sex and age-comparable subjects as a control group. Study population underwent metabolic characterization and carotid ultrasound for intima-media thickness measurement. Plasma OPN, OPG and OC were measured by MILLIPLEX Multiplex Assays Luminex. RESULTS T2DM patients had significantly higher circulating OPN and OPG levels than controls (14.3 ± 13.6 vs 10.6 ± 13.7 ng/ml p < 0.001, 0.70 ± 0.60 vs 0.54 ± 4.1 ng/ml, p = 0.02) while OC levels were similar in the two cohorts (6.35 ± 5.8 vs 7.80 ± 7.0 ng/ml, p = n.s). OPN and OPG positively correlated with greater systolic blood pressure (SBP) values, HOMA-IR and HOMA-β, and with the presence of dyslipidemia and carotid atherosclerosis. The association between greater OPN and OPG levels and SBP was independent from possible confounders (both p = 0.01). CONCLUSIONS Circulating OPN and OPG levels are increased in T2DM patients and identify a particularly unfavourable metabolic profile, mostly expressed by higher SBP. Bone peptides may represent novel markers of vascular stress and accelerated atherosclerosis in diabetes, constituting a possible tool for cardiovascular risk stratification in diabetes.
Collapse
Affiliation(s)
- I Barchetta
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - V Ceccarelli
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - F A Cimini
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - L Bertoccini
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - A Fraioli
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - C Alessandri
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - M G Baroni
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - M G Cavallo
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
25
|
Patel JJ, Bourne LE, Davies BK, Arnett TR, MacRae VE, Wheeler-Jones CP, Orriss IR. Differing calcification processes in cultured vascular smooth muscle cells and osteoblasts. Exp Cell Res 2019; 380:100-113. [PMID: 31004580 PMCID: PMC6520648 DOI: 10.1016/j.yexcr.2019.04.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 11/15/2022]
Abstract
Arterial medial calcification (AMC) is the deposition of calcium phosphate mineral, often as hydroxyapatite, in the medial layer of the arteries. AMC shares some similarities to skeletal mineralisation and has been associated with the transdifferentiation of vascular smooth muscle cells (VSMCs) towards an osteoblast-like phenotype. This study used primary mouse VSMCs and calvarial osteoblasts to directly compare the established and widely used in vitro models of AMC and bone formation. Significant differences were identified between osteoblasts and calcifying VSMCs. First, osteoblasts formed large mineralised bone nodules that were associated with widespread deposition of an extracellular collagenous matrix. In contrast, VSMCs formed small discrete regions of calcification that were not associated with collagen deposition and did not resemble bone. Second, calcifying VSMCs displayed a progressive reduction in cell viability over time (≤7-fold), with a 50% increase in apoptosis, whereas osteoblast and control VSMCs viability remained unchanged. Third, osteoblasts expressed high levels of alkaline phosphatase (TNAP) activity and TNAP inhibition reduced bone formation by to 90%. TNAP activity in calcifying VSMCs was ∼100-fold lower than that of bone-forming osteoblasts and cultures treated with β-glycerophosphate, a TNAP substrate, did not calcify. Furthermore, TNAP inhibition had no effect on VSMC calcification. Although, VSMC calcification was associated with increased mRNA expression of osteoblast-related genes (e.g. Runx2, osterix, osteocalcin, osteopontin), the relative expression of these genes was up to 40-fold lower in calcifying VSMCs versus bone-forming osteoblasts. In summary, calcifying VSMCs in vitro display some limited osteoblast-like characteristics but also differ in several key respects: 1) their inability to form collagen-containing bone; 2) their lack of reliance on TNAP to promote mineral deposition; and, 3) the deleterious effect of calcification on their viability.
Collapse
Affiliation(s)
- Jessal J Patel
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK; School of Life & Medical Sciences, University of Hertfordshire, Hatfield, UK
| | - Lucie E Bourne
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Bethan K Davies
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Vicky E MacRae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK.
| |
Collapse
|
26
|
Hypermethylation of the Micro-RNA 145 Promoter Is the Key Regulator for NLRP3 Inflammasome-Induced Activation and Plaque Formation. JACC Basic Transl Sci 2018; 3:604-624. [PMID: 30456333 PMCID: PMC6234615 DOI: 10.1016/j.jacbts.2018.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/14/2018] [Accepted: 06/19/2018] [Indexed: 01/17/2023]
Abstract
miR-145 in vessels decreases with plaque progression. DNMT1 and TET2 dynamic imbalance leads to miR-145 promoter hypermethylation. Reduction of miR-145 activates NLRP3 inflammasome through CD137/NFATc1 signaling. DNMT1 and TET2 could be promising therapeutic candidates for atherosclerosis in the future.
Two major issues are involved in clinical atherosclerosis treatment. First, there are no significant clinical markers for early diagnosis of atherosclerosis. Second, the plaque will not regress once it initiates even if the risk factors are removed. In this paper, the research shows that the hypermethylation level of the microRNA 145 (miR-145) promoter is related to a DNMT1 and TET2 dynamic imbalance. The reduction of miR-145 causes NLRP3 (nucleotide-binding oligomerization domain-like receptor protein 3) inflammasome activation through CD137/NFATc1 signaling. These findings could be a potential target for plaque regression in the future.
Collapse
|
27
|
Skafi N, Abdallah D, Soulage C, Reibel S, Vitale N, Hamade E, Faour W, Magne D, Badran B, Hussein N, Buchet R, Brizuela L, Mebarek S. Phospholipase D: A new mediator during high phosphate-induced vascular calcification associated with chronic kidney disease. J Cell Physiol 2018; 234:4825-4839. [PMID: 30207376 DOI: 10.1002/jcp.27281] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/26/2018] [Indexed: 01/31/2023]
Abstract
Vascular calcification (VC) is the pathological accumulation of calcium phosphate crystals in one of the layers of blood vessels, leading to loss of elasticity and causing severe calcification in vessels. Medial calcification is mostly seen in patients with chronic kidney disease (CKD) and diabetes. Identification of key enzymes and their actions during calcification will contribute to understand the onset of pathological calcification. Phospholipase D (PLD1, PLD2) is active at the earlier steps of mineralization in osteoblasts and chondrocytes. In this study, we aimed to determine their effects during high-phosphate treatment in mouse vascular smooth muscle cell line MOVAS, in the ex vivo model of the rat aorta, and in the in vivo model of adenine-induced CKD. We observed an early increase in PLD1 gene and protein expression along with the increase in the PLD activity in vascular muscle cell line, during calcification induced by ascorbic acid and β-glycerophosphate. Inhibition of PLD1 by the selective inhibitor VU0155069, or the pan-PLD inhibitor, halopemide, prevented calcification. The mechanism of PLD activation is likely to be protein kinase C (PKC)-independent since bisindolylmaleimide X hydrochloride, a pan-PKC inhibitor, did not affect the PLD activity. In agreement, we found an increase in Pld1 gene expression and PLD activity in aortic explant cultures treated with high phosphate, whereas PLD inhibition by halopemide decreased calcification. Finally, an increase in both Pld1 and Pld2 expression occurred simultaneously with the appearance of VC in a rat model of CKD. Thus, PLD, especially PLD1, promotes VC in the context of CKD and could be an important target for preventing onset or progression of VC.
Collapse
Affiliation(s)
- Najwa Skafi
- University of Lyon, Université Claude Bernard Lyon 1 (UCBL), CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), Lyon, France.,Genomic and Health Laboratory/PRASE-EDST Campus Rafic Hariri-Hadath-Beirut-Liban, Faculty of Sciences, Lebanese University (LU), Beirut, Lebanon
| | - Dina Abdallah
- University of Lyon, Université Claude Bernard Lyon 1 (UCBL), CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), Lyon, France.,Genomic and Health Laboratory/PRASE-EDST Campus Rafic Hariri-Hadath-Beirut-Liban, Faculty of Sciences, Lebanese University (LU), Beirut, Lebanon
| | - Christophe Soulage
- University of Lyon, CarMeN, INSERM U1060, INRA U1397, Institut National des Sciences Appliquées de Lyon (INSA Lyon), Université Claude Bernard Lyon 1, Villeurbanne, France
| | | | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives (INCI), UPR-3212 CNRS and Université de Strasbourg, Strasbourg, France
| | - Eva Hamade
- Genomic and Health Laboratory/PRASE-EDST Campus Rafic Hariri-Hadath-Beirut-Liban, Faculty of Sciences, Lebanese University (LU), Beirut, Lebanon
| | - Wissam Faour
- School of Medicine, Lebanese American University (LAU), Byblos, Lebanon
| | - David Magne
- University of Lyon, Université Claude Bernard Lyon 1 (UCBL), CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), Lyon, France
| | - Bassam Badran
- Genomic and Health Laboratory/PRASE-EDST Campus Rafic Hariri-Hadath-Beirut-Liban, Faculty of Sciences, Lebanese University (LU), Beirut, Lebanon
| | - Nader Hussein
- Genomic and Health Laboratory/PRASE-EDST Campus Rafic Hariri-Hadath-Beirut-Liban, Faculty of Sciences, Lebanese University (LU), Beirut, Lebanon
| | - Rene Buchet
- University of Lyon, Université Claude Bernard Lyon 1 (UCBL), CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), Lyon, France
| | - Leyre Brizuela
- University of Lyon, Université Claude Bernard Lyon 1 (UCBL), CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), Lyon, France
| | - Saida Mebarek
- University of Lyon, Université Claude Bernard Lyon 1 (UCBL), CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), Lyon, France
| |
Collapse
|
28
|
Roszkowska M, Strzelecka-Kiliszek A, Bessueille L, Buchet R, Magne D, Pikula S. Collagen promotes matrix vesicle-mediated mineralization by vascular smooth muscle cells. J Inorg Biochem 2018; 186:1-9. [DOI: 10.1016/j.jinorgbio.2018.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/20/2018] [Accepted: 05/16/2018] [Indexed: 02/08/2023]
|
29
|
The Bioactive Substance Secreted by MSC Retards Mouse Aortic Vascular Smooth Muscle Cells Calcification. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6053567. [PMID: 29967775 PMCID: PMC6008760 DOI: 10.1155/2018/6053567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/07/2018] [Indexed: 12/13/2022]
Abstract
Background Vascular calcification, which is associated with low-level chronic inflammation, is a complication that occurs during aging, atherosclerosis, chronic kidney disease, diabetes mellitus, and hyperlipaemia. In this study, we used conditioned media from mesenchymal stem cells (MSC-CM), a source of autologous cytokines, to test the hypothesis that MSC-CM inhibits vascular smooth muscle cell (VSMC) calcification by suppressing inflammation and apoptosis. Methods VSMCs were treated with β-glycerophosphate (β-GP) to induce calcification and MSC-CM was used as a treatment. Calcium deposition was evaluated using alizarin red and von Kossa staining after a 7-day induction period. Intracellular calcium contents were measured via the o-cresolphthalein complexone method, and alkaline phosphatase (ALP) activity was determined using the para-nitrophenyl phosphate method. The expressions of specific-osteogenic markers, inflammatory cytokines, and apoptosis-associated genes/proteins were examined by real-time polymerase chain reaction or western blotting. Results MSC-CM inhibited β-GP-induced calcium deposition in VSMCs and decreased intracellular calcium content and ALP activity. Additionally, MSC-CM suppressed the β-GP-induced increases in BMP2, Msx2, Runx2, and osteocalcin expression. Additionally, MSC-CM decreased the expression of TNF-α, IL-1β, and IL-6 in VSMC. MSC-CM also partly blocked β-GP-induced VSMC apoptosis, which was associated with an increase in the Bcl-2/Bax expression ratio and a decrease in caspase-3 expression. Conclusion Our study results suggest that MSC-CM can inhibit VSMC calcification. This suggests a potential novel clinical application for MSCs in the treatment of vascular calcification and associated diseases.
Collapse
|
30
|
Schwarz S, Mrosewski I, Silawal S, Schulze-Tanzil G. The interrelation of osteoarthritis and diabetes mellitus: considering the potential role of interleukin-10 and in vitro models for further analysis. Inflamm Res 2017; 67:285-300. [PMID: 29196771 DOI: 10.1007/s00011-017-1121-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 11/12/2017] [Accepted: 11/24/2017] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Today, not only the existence of an interrelation between obesity/adipositas and osteoarthritis (OA) but also the association of OA and diabetes mellitus (DM) are widely recognized. Nevertheless, shared influence factors facilitating OA development in DM patients still remain speculative up until now. To supplement the analysis of clinical data, appropriate in vitro models could help to identify shared pathogenetic pathways. Informative in vitro studies could later be complemented by in vivo data obtained from suitable animal models. MATERIALS AND METHODS Therefore, this detailed review of available literature was undertaken to discuss and compare the results of currently published in vitro studies focusing on the interrelation between OA, the metabolic syndrome and DM and to propose models to further study the molecular pathways. RESULTS The survey of literature presented here supports the hypothesis that the pathogenesis of OA in DM is based on imbalanced molecular pathways with a putative crucial role of antiinflammatory cytokines such as IL-10. CONCLUSION Future development of versatile micro-scaled in vitro models such as combining DM and OA on chip could allow the identification of common pathogenetic pathways and might help to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Silke Schwarz
- Department of Anatomy, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany.,Institute of Anatomy, Paracelsus Medical University, Salzburg, Austria
| | - Ingo Mrosewski
- MVZ Limbach Laboratories, Aroser Allee 84, 13407, Berlin, Germany
| | - Sandeep Silawal
- Department of Anatomy, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany.,Institute of Anatomy, Paracelsus Medical University, Salzburg, Austria
| | - Gundula Schulze-Tanzil
- Department of Anatomy, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, 90419, Nuremberg, Germany. .,Institute of Anatomy, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
31
|
Fakhry M, Roszkowska M, Briolay A, Bougault C, Guignandon A, Diaz-Hernandez JI, Diaz-Hernandez M, Pikula S, Buchet R, Hamade E, Badran B, Bessueille L, Magne D. TNAP stimulates vascular smooth muscle cell trans-differentiation into chondrocytes through calcium deposition and BMP-2 activation: Possible implication in atherosclerotic plaque stability. Biochim Biophys Acta Mol Basis Dis 2016; 1863:643-653. [PMID: 27932058 DOI: 10.1016/j.bbadis.2016.12.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/12/2016] [Accepted: 12/04/2016] [Indexed: 01/15/2023]
Abstract
Atherosclerotic plaque calcification varies from early, diffuse microcalcifications to a bone-like tissue formed by endochondral ossification. Recently, a paradigm has emerged suggesting that if the bone metaplasia stabilizes the plaques, microcalcifications are harmful. Tissue-nonspecific alkaline phosphatase (TNAP), an ectoenzyme necessary for mineralization by its ability to hydrolyze inorganic pyrophosphate (PPi), is stimulated by inflammation in vascular smooth muscle cells (VSMCs). Our objective was to determine the role of TNAP in trans-differentiation of VSMCs and calcification. In rodent MOVAS and A7R5 VSMCs, addition of exogenous alkaline phosphatase (AP) or TNAP overexpression was sufficient to stimulate the expression of several chondrocyte markers and induce mineralization. Addition of exogenous AP to human mesenchymal stem cells cultured in pellets also stimulated chondrogenesis. Moreover, TNAP inhibition with levamisole in mouse primary chondrocytes dropped mineralization as well as the expression of chondrocyte markers. VSMCs trans-differentiated into chondrocyte-like cells, as well as primary chondrocytes, used TNAP to hydrolyze PPi, and PPi provoked the same effects as TNAP inhibition in primary chondrocytes. Interestingly, apatite crystals, associated or not to collagen, mimicked the effects of TNAP on VSMC trans-differentiation. AP and apatite crystals increased the expression of BMP-2 in VSMCs, and TNAP inhibition reduced BMP-2 levels in chondrocytes. Finally, the BMP-2 inhibitor noggin blocked the rise in aggrecan induced by AP in VSMCs, suggesting that TNAP induction in VSMCs triggers calcification, which stimulates chondrogenesis through BMP-2. Endochondral ossification in atherosclerotic plaques may therefore be induced by crystals, probably to confer stability to plaques with microcalcifications.
Collapse
Affiliation(s)
- Maya Fakhry
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France; Lebanese University, Laboratory of Cancer Biology and Molecular Immunology, EDST-PRASE, Hadath-Beirut, Lebanon
| | - Monika Roszkowska
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France; Laboratory of Biochemistry of Lipids, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Anne Briolay
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - Carole Bougault
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - Alain Guignandon
- Univ Lyon, Université Jean Monnet Saint-Etienne, LBTO, UMR INSERM 1059, F-42023 Saint-Etienne, France
| | - Juan Ignacio Diaz-Hernandez
- Universidad Complutense de Madrid, Facultad de Veterinaria, Dpt. Bioquimica y Biologia Molecular IV, Madrid, Spain
| | - Miguel Diaz-Hernandez
- Universidad Complutense de Madrid, Facultad de Veterinaria, Dpt. Bioquimica y Biologia Molecular IV, Madrid, Spain
| | - Slawomir Pikula
- Laboratory of Biochemistry of Lipids, Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - René Buchet
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France
| | - Eva Hamade
- Lebanese University, Laboratory of Cancer Biology and Molecular Immunology, EDST-PRASE, Hadath-Beirut, Lebanon
| | - Bassam Badran
- Lebanese University, Laboratory of Cancer Biology and Molecular Immunology, EDST-PRASE, Hadath-Beirut, Lebanon
| | | | - David Magne
- Univ Lyon, University Lyon 1, ICBMS, UMR CNRS 5246, F-69622 Lyon, France.
| |
Collapse
|
32
|
Harper E, Forde H, Davenport C, Rochfort KD, Smith D, Cummins PM. Vascular calcification in type-2 diabetes and cardiovascular disease: Integrative roles for OPG, RANKL and TRAIL. Vascul Pharmacol 2016; 82:30-40. [DOI: 10.1016/j.vph.2016.02.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/01/2016] [Accepted: 02/21/2016] [Indexed: 12/14/2022]
|