1
|
Tóth A, Balogh E, Jeney V. In Vitro Models of Cardiovascular Calcification. Biomedicines 2024; 12:2155. [PMID: 39335668 PMCID: PMC11429067 DOI: 10.3390/biomedicines12092155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/15/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiovascular calcification, characterized by hydroxyapatite deposition in the arterial wall and heart valves, is associated with high cardiovascular morbidity and mortality. Cardiovascular calcification is a hallmark of aging but is frequently seen in association with chronic diseases, such as chronic kidney disease (CKD), diabetes, dyslipidemia, and hypertension in the younger population as well. Currently, there is no therapeutic approach to prevent or cure cardiovascular calcification. The pathophysiology of cardiovascular calcification is highly complex and involves osteogenic differentiation of various cell types of the cardiovascular system, such as vascular smooth muscle cells and valve interstitial cells. In vitro cellular and ex vivo tissue culture models are simple and useful tools in cardiovascular calcification research. These models contributed largely to the discoveries of the numerous calcification inducers, inhibitors, and molecular mechanisms. In this review, we provide an overview of the in vitro cell culture and the ex vivo tissue culture models applied in the research of cardiovascular calcification.
Collapse
Affiliation(s)
- Andrea Tóth
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Enikő Balogh
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Viktória Jeney
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
2
|
Molinuevo MS, Cortizo AM, Sedlinsky C. Effects of advanced glycation end-products, diabetes and metformin on the osteoblastic transdifferentiation capacity of vascular smooth muscle cells: In vivo and in vitro studies. J Diabetes Complications 2023; 37:108626. [PMID: 37839167 DOI: 10.1016/j.jdiacomp.2023.108626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/27/2023] [Accepted: 10/02/2023] [Indexed: 10/17/2023]
Abstract
AIMS Our objective was to study the vascular smooth muscle cells (VSMC) osteoblastic transdifferentiation in AGE exposed cells or those from diabetic animals, and its response to metformin treatment. METHODS VSMC were obtained from non-diabetic rats, grown with or without AGE; while VSMC of in vivo-ex vivo studies were obtained from non-diabetic control animals (C), diabetic (D), C treated with metformin (M) and D treated with metformin (D-M). We studied the osteoblastic differentiation by evaluating alkaline phosphatase (ALP), type I collagen (Col) and mineral deposit. RESULTS In vitro, AGE increased proliferation, migration, and osteoblastic differentiation of VSMC. Metformin cotreatment prevented the AGE induced proliferation and migration. Both AGE and metformin stimulated the expression of ALP and Col. AGE induced mineralization was prevented by metformin. VSMC from D expressed a higher production of Col and ALP. Those from D-M showed an ALP increase vs C and M, and a partial decrease vs D. Cultured in osteogenic medium, ALP, Col and mineralization increased in D vs C, remained unchanged in M, and were prevented in D-M animals. CONCLUSION Both AGE and DM favor VSMC differentiation towards the osteogenic phenotype and this effect can be prevented by metformin.
Collapse
Affiliation(s)
- María Silvina Molinuevo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral UNLP-CICPBA, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 50 y 115, 1900 La Plata, Argentina
| | - Ana María Cortizo
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral UNLP-CICPBA, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 50 y 115, 1900 La Plata, Argentina.
| | - Claudia Sedlinsky
- Laboratorio de Investigaciones en Osteopatías y Metabolismo Mineral UNLP-CICPBA, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, 50 y 115, 1900 La Plata, Argentina.
| |
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
|
Zhang YR, Liu SM, Chen Y, Zhang LS, Ji DR, Zhao J, Yu YR, Jia MZ, Tang CS, Huang W, Zhou YB, Chai SB, Qi YF. Intermedin alleviates diabetic vascular calcification by inhibiting GLUT1 through activation of the cAMP/PKA signaling pathway. Atherosclerosis 2023; 385:117342. [PMID: 37879153 DOI: 10.1016/j.atherosclerosis.2023.117342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/21/2023] [Accepted: 10/10/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND AND AIMS Vascular calcification (VC) is regarded as an independent risk factor for cardiovascular events in type 2 diabetic patients. Glucose transporter 1 (GLUT1) involves VC. Intermedin/Adrenomedullin-2 (IMD/ADM2) is a cardiovascular protective peptide that can inhibit multiple disease-associated VC. However, the role and mechanism of IMD in diabetic VC remain unclear. Here, we investigated whether IMD inhibits diabetic VC by inhibiting GLUT1. METHODS AND RESULTS It was found that plasma IMD concentration was significantly decreased in type 2 diabetic patients and in fructose-induced diabetic rats compared with that in controls. Plasma IMD content was inversely correlated with fasting blood glucose level and VC severity. IMD alleviated VC in fructose-induced diabetic rats. Deficiency of Adm2 aggravated and Adm2 overexpression attenuated VC in high-fat diet-induced diabetic mice. In vitro, IMD mitigated high glucose-induced calcification of vascular smooth muscle cells (VSMCs). Mechanistically, IMD reduced advanced glycation end products (AGEs) content and the level of receptor for AGEs (RAGE). IMD decreased glucose transporter 1 (GLUT1) levels. The inhibitory effect of IMD on RAGE protein level was blocked by GLUT1 knockdown. GLUT1 knockdown abolished the effect of IMD on alleviating VSMC calcification. IMD receptor antagonist IMD17-47 and cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) inhibitor H89 abolished the inhibitory effects of IMD on GLUT1 and VSMC calcification. CONCLUSIONS These findings revealed that IMD exerted its anti-calcification effect by inhibiting GLUT1, providing a novel therapeutic target for diabetic VC.
Collapse
Affiliation(s)
- Ya-Rong Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Shi-Meng Liu
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Yao Chen
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Lin-Shuang Zhang
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Deng-Ren Ji
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Jie Zhao
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Yan-Rong Yu
- Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Mo-Zhi Jia
- Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China
| | - Chao-Shu Tang
- StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China
| | - Wei Huang
- StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China
| | - Ye-Bo Zhou
- Department of Physiology, Nanjing Medical University, Nanjing, 211166, China.
| | - San-Bao Chai
- Department of Endocrinology and Metabolism, Peking University International Hospital, Beijing, 102206, China.
| | - Yong-Fen Qi
- Laboratory of Cardiovascular Bioactive Molecule, School of Basic Medical Sciences, Peking University, Beijing, 100083, China; StateKey Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100083, China; Department of Pathogen Biology, School of Basic Medical Science, Peking University, Beijing, 100083, China.
| |
Collapse
|
5
|
Ishikawa M, Kanzaki H, Kodera R, Sekimizu T, Wada S, Tohyama S, Ida T, Shimoyama M, Manase S, Tomonari H, Kuroda N. Early diagnosis of aortic calcification through dental X-ray examination for dental pulp stones. Sci Rep 2023; 13:18576. [PMID: 37903847 PMCID: PMC10616172 DOI: 10.1038/s41598-023-45902-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/25/2023] [Indexed: 11/01/2023] Open
Abstract
Vascular calcification, an ectopic calcification exacerbated by aging and renal dysfunction, is closely associated with cardiovascular disease. However, early detection indicators are limited. This study focused on dental pulp stones, ectopic calcifications found in oral tissues that are easily identifiable on dental radiographs. Our investigation explored the frequency and timing of these calcifications in different locations and their relationship to aortic calcification. In cadavers, we examined the association between the frequency of dental pulp stones and aortic calcification, revealing a significant association. Notably, dental pulp stones appeared prior to aortic calcification. Using a rat model of hyperphosphatemia, we confirmed that dental pulp stones formed earlier than calcification in the aortic arch. Interestingly, there were very few instances of aortic calcification without dental pulp stones. Additionally, we conducted cell culture experiments with vascular smooth muscle cells (SMCs) and dental pulp cells (DPCs) to explore the regulatory mechanism underlying high phosphate-mediated calcification. We found that DPCs produced calcification deposits more rapidly and exhibited a stronger augmentation of osteoblast differentiation markers compared with SMCs. In conclusion, the observation of dental pulp stones through X-ray examination during dental checkups could be a valuable method for early diagnosis of aortic calcification risk.
Collapse
Affiliation(s)
- Misao Ishikawa
- Department of Anatomy, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, Kanagawa Pref., 230-8501, Japan.
| | - Hiroyuki Kanzaki
- Department of Orthodontics, School of Dental Medicine, Tsurumi University, Yokohama, Japan
| | - Ryo Kodera
- Department of Anatomy, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, Kanagawa Pref., 230-8501, Japan
| | - Takehiro Sekimizu
- Department of Anatomy, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, Kanagawa Pref., 230-8501, Japan
| | - Satoshi Wada
- Department of Oral and Maxillofacial Surgery, Kanazawa Medical University, Kanazawa, Japan
| | - Syunnosuke Tohyama
- Department of Orthodontics, School of Dental Medicine, Tsurumi University, Yokohama, Japan
| | - Tomomi Ida
- Department of Orthodontics, School of Dental Medicine, Tsurumi University, Yokohama, Japan
| | - Miho Shimoyama
- Department of Orthodontics, School of Dental Medicine, Tsurumi University, Yokohama, Japan
| | - Shugo Manase
- Department of Orthodontics, School of Dental Medicine, Tsurumi University, Yokohama, Japan
| | - Hiroshi Tomonari
- Department of Orthodontics, School of Dental Medicine, Tsurumi University, Yokohama, Japan
| | - Noriyuki Kuroda
- Department of Anatomy, School of Dental Medicine, Tsurumi University, 2-1-3 Tsurumi, Tsurumi-ku, Yokohama, Kanagawa Pref., 230-8501, Japan
| |
Collapse
|
6
|
Abstract
The medial layer of the arterial wall is composed mainly of vascular smooth muscle cells (VSMCs). Under physiological conditions, VSMCs assume a contractile phenotype, and their primary function is to regulate vascular tone. In contrast with terminally differentiated cells, VSMCs possess phenotypic plasticity, capable of transitioning into other cellular phenotypes in response to changes in the vascular environment. Recent research has shown that VSMC phenotypic switching participates in the pathogenesis of atherosclerosis, where the various types of dedifferentiated VSMCs accumulate in the atherosclerotic lesion and participate in the associated vascular remodeling by secreting extracellular matrix proteins and proteases. This review article discusses the 9 VSMC phenotypes that have been reported in atherosclerotic lesions and classifies them into differentiated VSMCs, intermediately dedifferentiated VSMCs, and dedifferentiated VSMCs. It also provides an overview of several methodologies that have been developed for studying VSMC phenotypic switching and discusses their respective advantages and limitations.
Collapse
Affiliation(s)
- Runji Chen
- Shantou University Medical CollegeShantouChina
| | - David G. McVey
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUnited Kingdom
| | - Daifei Shen
- Research Center for Translational MedicineThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | | | - Shu Ye
- Shantou University Medical CollegeShantouChina
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUnited Kingdom
- Cardiovascular‐Metabolic Disease Translational Research ProgrammeNational University of SingaporeSingapore
| |
Collapse
|
7
|
Moradi L, Witek L, Vivekanand Nayak V, Cabrera Pereira A, Kim E, Good J, Liu CJ. Injectable hydrogel for sustained delivery of progranulin derivative Atsttrin in treating diabetic fracture healing. Biomaterials 2023; 301:122289. [PMID: 37639975 PMCID: PMC11232488 DOI: 10.1016/j.biomaterials.2023.122289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/22/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
Hydrogels with long-term storage stability, controllable sustained-release properties, and biocompatibility have been garnering attention as carriers for drug/growth factor delivery in tissue engineering applications. Chitosan (CS)/Graphene Oxide (GO)/Hydroxyethyl cellulose (HEC)/β-glycerol phosphate (β-GP) hydrogel is capable of forming a 3D gel network at physiological temperature (37 °C), rendering it an excellent candidate for use as an injectable biomaterial. This work focused on an injectable thermo-responsive CS/GO/HEC/β-GP hydrogel, which was designed to deliver Atsttrin, an engineered derivative of a known chondrogenic and anti-inflammatory growth factor-like molecule progranulin. The combination of the CS/GO/HEC/β-GP hydrogel and Atsttrin provides a unique biochemical and biomechanical environment to enhance fracture healing. CS/GO/HEC/β-GP hydrogels with increased amounts of GO exhibited rapid sol-gel transition, higher viscosity, and sustained release of Atsttrin. In addition, these hydrogels exhibited a porous interconnected structure. The combination of Atsttrin and hydrogel successfully promoted chondrogenesis and osteogenesis of bone marrow mesenchymal stem cells (bmMSCs) in vitro. Furthermore, the work also presented in vivo evidence that injection of Atsttrin-loaded CS/GO/HEC/β-GP hydrogel stimulated diabetic fracture healing by simultaneously inhibiting inflammatory and stimulating cartilage regeneration and endochondral bone formation signaling pathways. Collectively, the developed injectable thermo-responsive CS/GO/HEC/βG-P hydrogel yielded to be minimally invasive, as well as capable of prolonged and sustained delivery of Atsttrin, for therapeutic application in impaired fracture healing, particularly diabetic fracture healing.
Collapse
Affiliation(s)
- Lida Moradi
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA; Department of Orthopaedics & Rehabilitation, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Lukasz Witek
- Biomaterials Division - Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA; Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY, 11201, USA
| | - Vasudev Vivekanand Nayak
- Biomaterials Division - Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Angel Cabrera Pereira
- Biomaterials Division - Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Ellen Kim
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA
| | - Julia Good
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics Surgery, New York University Grossman School of Medicine, New York, NY, 10003, USA; Department of Orthopaedics & Rehabilitation, Yale University School of Medicine, New Haven, CT, 06510, USA; Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
8
|
Hypoxia-inducible factor signaling in vascular calcification in chronic kidney disease patients. J Nephrol 2022; 35:2205-2213. [PMID: 36208406 DOI: 10.1007/s40620-022-01432-8] [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: 03/11/2022] [Accepted: 08/02/2022] [Indexed: 10/10/2022]
Abstract
Chronic kidney disease (CKD) affects approximately 15% of the adult population in high-income countries and is associated with significant comorbidities, including increased vascular calcifications which is associated with a higher risk for cardiovascular events. Even though the underlying pathophysiology is unclear, hypoxia-inducible factor (HIF) signaling appears to play a central role in inflammation, angiogenesis, fibrosis, cellular proliferation, apoptosis and vascular calcifications which is influenced by multiple variables such as iron deficiency anemia, serum phosphorus and calcium levels, fibroblast growth factor-23 (FGF-23) and Klotho. Along with the growing understanding of the pathology, potential therapeutic alternatives have emerged including HIF stabilizers and SGLT-2 inhibitors. The aim of this review is to discuss the role of HIF signaling in the pathophysiology of vascular calcification in CKD patients and to identify potential therapeutic approaches.
Collapse
|
9
|
Kamato D, Ilyas I, Xu S, Little PJ. Non-Mouse Models of Atherosclerosis: Approaches to Exploring the Translational Potential of New Therapies. Int J Mol Sci 2022; 23:12964. [PMID: 36361754 PMCID: PMC9656683 DOI: 10.3390/ijms232112964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 09/26/2023] Open
Abstract
Cardiovascular disease is the largest single cause of disease-related mortality worldwide and the major underlying pathology is atherosclerosis. Atherosclerosis develops as a complex process of vascular lipid deposition and retention by modified proteoglycans, endothelial dysfunction and unresolved chronic inflammation. There are a multitude of current therapeutic agents, most based on lowering plasma lipid levels, but, overall, they have a lower than optimum level of efficacy and many deaths continue to arise from cardiovascular disease world-wide. To identify and evaluate potential novel cardiovascular drugs, suitable animal models that reproduce human atherosclerosis with a high degree of fidelity are required as essential pre-clinical research tools. Commonly used animal models of atherosclerosis include mice (ApoE-/-, LDLR-/- mice and others), rabbits (WHHL rabbits and others), rats, pigs, hamster, zebrafish and non-human primates. Models based on various wild-type and genetically modified mice have been extensively reviewed but mice may not always be appropriate. Thus, here, we provide an overview of the advantages and shortcomings of various non-mouse animal models of atherosclerotic plaque formation, and plaque rupture, as well as commonly used interventional strategies. Taken together, the combinatorial selection of suitable animal models readily facilitates reproducible and rigorous translational research in discovering and validating novel anti-atherosclerotic drugs.
Collapse
Affiliation(s)
- Danielle Kamato
- Discovery Biology, Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
- Pharmacy Australia Centre of Excellence, School of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Iqra Ilyas
- Laboratory of Metabolics and Cardiovascular Diseases, University of Science and Technology of China, Hefei 230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
| | - Suowen Xu
- Laboratory of Metabolics and Cardiovascular Diseases, University of Science and Technology of China, Hefei 230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei 230001, China
| | - Peter J. Little
- Pharmacy Australia Centre of Excellence, School of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
- Sunshine Coast Health Institute and School of Health and Behavioural Sciences, University of the Sunshine Coast, Birtinya, QLD 4575, Australia
| |
Collapse
|
10
|
Negri AL. Role of prolyl hydroxylase/HIF-1 signaling in vascular calcification. Clin Kidney J 2022; 16:205-209. [PMID: 36755843 PMCID: PMC9900523 DOI: 10.1093/ckj/sfac224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Morbidity and mortality of chronic kidney disease (CKD) patients are largely associated with vascular calcification, an actively regulated process in which vascular smooth muscle cells (VSMCs) change into cells similar to osteocytes/chondrocytes, known as trans-differentiation. Cellular and systemic response to low oxygen (hypoxia) is regulated by the prolyl hydroxylase/hypoxia-inducible factor (HIF)-1 pathway. Recent studies highlighted that hypoxia-mediated activation of HIF-1 induces trans-differentiation of VSMCs into bone-forming type through an increase in osteo-/chondrogenic genes. Inhibition of the HIF-1 pathway abolished osteochondrogenic differentiation of VSMCs. Hypoxia strongly enhanced elevated phosphate-induced VSMC osteogenic trans-differentiation and calcification. HIF-1 was shown to be essential for phosphate enhanced VSMC calcification. O2-dependent degradation HIF-1 is triggered by the prolyl hydroxylase domain proteins (PHD). Prolyl hydroxylase inhibitors, daprodustat and roxadustat, increase high phosphate-induced VC in VSMCs, stabilizing HIF-1α and activating the HIF-1 pathway in these cells. Whether the use of these PHD inhibitors to treat anemia in CKD patients will favor the development and progression of vascular calcification remains to be explored.
Collapse
|
11
|
Ceccherini E, Cecchettini A, Gisone I, Persiani E, Morales MA, Vozzi F. Vascular Calcification: In Vitro Models under the Magnifying Glass. Biomedicines 2022; 10:biomedicines10102491. [PMID: 36289753 DOI: 10.3390/biomedicines10102491] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular calcification is a systemic disease contributing to cardiovascular morbidity and mortality. The pathophysiology of vascular calcification involves calcium salt deposition by vascular smooth muscle cells that exhibit an osteoblast-like phenotype. Multiple conditions drive the phenotypic switch and calcium deposition in the vascular wall; however, the exact molecular mechanisms and the connection between vascular smooth muscle cells and other cell types are not fully elucidated. In this hazy landscape, effective treatment options are lacking. Due to the pathophysiological complexity, several research models are available to evaluate different aspects of the calcification process. This review gives an overview of the in vitro cell models used so far to study the molecular processes underlying vascular calcification. In addition, relevant natural and synthetic compounds that exerted anticalcifying properties in in vitro systems are discussed.
Collapse
Affiliation(s)
- Elisa Ceccherini
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Antonella Cecchettini
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Ilaria Gisone
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Elisa Persiani
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Maria Aurora Morales
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Federico Vozzi
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| |
Collapse
|
12
|
Attenuating effect of magnesium on pulmonary arterial calcification in rodent models of pulmonary hypertension. J Hypertens 2022; 40:1979-1993. [PMID: 36052522 DOI: 10.1097/hjh.0000000000003211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Vascular calcification has been considered as a potential therapeutic target in pulmonary hypertension. Mg2+ has a protective role against calcification. This study aimed to investigate whether Mg2+ could alleviate pulmonary hypertension by reducing medial calcification of pulmonary arteries. METHODS Monocrotaline (MCT)-induced and chronic hypoxia-induced pulmonary hypertension rats were given an oral administration of 10% MgSO4 (10 ml/kg per day). Additionally, we administered Mg2+ in calcified pulmonary artery smooth muscle cells (PASMCs) after incubating with β-glycerophosphate (β-GP, 10 mmol/l). RESULTS In vivo, MCT-induced and chronic hypoxia-induced pulmonary hypertension indexes, including right ventricular systolic pressure, right ventricular mass index, and arterial wall thickness, as well as Alizarin Red S (ARS) staining-visualized calcium deposition, high calcium levels, and osteochondrogenic differentiation in pulmonary arteries, were mitigated by dietary Mg2+ intake. In vitro, β-GP-induced calcium-rich deposits stained by ARS, calcium content, as well as the detrimental effects of calcification to proliferation, migration, and resistance to apoptosis of PASMCs were alleviated by high Mg2+ but exacerbated by low Mg2+. Expression levels of mRNA and protein of β-GP-induced osteochondrogenic markers, RUNX Family Transcription Factor 2, and Msh Homeobox 2 were decreased by high Mg2+ but increased by low Mg2+; however, Mg2+ did not affect β-GP-induced expression of SRY-Box Transcription Factor 9. Moreover, mRNA expression and protein levels of β-GP-reduced calcification inhibitor, Matrix GLA protein was increased by high Mg2+ but decreased by low Mg2+. CONCLUSION Mg2+ supplement is a powerful strategy to treat pulmonary hypertension by mitigating pulmonary arterial calcification as the calcification triggered physiological and pathological changes to PASMCs.
Collapse
|
13
|
Heuschkel MA, Babler A, Heyn J, van der Vorst EPC, Steenman M, Gesper M, Kappel BA, Magne D, Gouëffic Y, Kramann R, Jahnen-Dechent W, Marx N, Quillard T, Goettsch C. Distinct role of mitochondrial function and protein kinase C in intimal and medial calcification in vitro. Front Cardiovasc Med 2022; 9:959457. [PMID: 36204585 PMCID: PMC9530266 DOI: 10.3389/fcvm.2022.959457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction Vascular calcification (VC) is a major risk factor for cardiovascular morbidity and mortality. Depending on the location of mineral deposition within the arterial wall, VC is classified as intimal and medial calcification. Using in vitro mineralization assays, we developed protocols triggering both types of calcification in vascular smooth muscle cells (SMCs) following diverging molecular pathways. Materials and methods and results Human coronary artery SMCs were cultured in osteogenic medium (OM) or high calcium phosphate medium (CaP) to induce a mineralized extracellular matrix. OM induces osteoblast-like differentiation of SMCs-a key process in intimal calcification during atherosclerotic plaque remodeling. CaP mimics hyperphosphatemia, associated with chronic kidney disease-a risk factor for medial calcification. Transcriptomic analysis revealed distinct gene expression profiles of OM and CaP-calcifying SMCs. OM and CaP-treated SMCs shared 107 differentially regulated genes related to SMC contraction and metabolism. Real-time extracellular efflux analysis demonstrated decreased mitochondrial respiration and glycolysis in CaP-treated SMCs compared to increased mitochondrial respiration without altered glycolysis in OM-treated SMCs. Subsequent kinome and in silico drug repurposing analysis (Connectivity Map) suggested a distinct role of protein kinase C (PKC). In vitro validation experiments demonstrated that the PKC activators prostratin and ingenol reduced calcification triggered by OM and promoted calcification triggered by CaP. Conclusion Our direct comparison results of two in vitro calcification models strengthen previous observations of distinct intracellular mechanisms that trigger OM and CaP-induced SMC calcification in vitro. We found a differential role of PKC in OM and CaP-calcified SMCs providing new potential cellular and molecular targets for pharmacological intervention in VC. Our data suggest that the field should limit the generalization of results found in in vitro studies using different calcification protocols.
Collapse
Affiliation(s)
- Marina A. Heuschkel
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Anne Babler
- Institute of Experimental Medicine and Systems Biology, University Hospital, RWTH Aachen, Aachen, Germany
| | - Jonas Heyn
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Emiel P. C. van der Vorst
- Interdisciplinary Center for Clinical Research, Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Marja Steenman
- L’institut Du Thorax, Inserm UMR 1087, CNRS, INSERM, France and Nantes Université, Nantes, France
| | - Maren Gesper
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Ben A. Kappel
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - David Magne
- ICBMS UMR CNRS 5246, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Yann Gouëffic
- Department of Vascular Surgery, Vascular Center, Groupe Hospitalier Paris Saint-Joseph, Paris, France
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology, University Hospital, RWTH Aachen, Aachen, Germany
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, Netherlands
| | - Willi Jahnen-Dechent
- Biointerface Laboratory, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Nikolaus Marx
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Thibaut Quillard
- L’institut Du Thorax, Inserm UMR 1087, CNRS, INSERM, France and Nantes Université, Nantes, France
- PHY-OS Laboratory, INSERM UMR 1238, Nantes University of Medicine, Nantes, France
| | - Claudia Goettsch
- Department of Internal Medicine I–Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
14
|
Zhang F, Li J, Gu C, Zhang H. MiR-140-5p upregulation suppressed β-glycerophosphate-induced vascular smooth muscle cell calcification via targeting TLR4. Immunopharmacol Immunotoxicol 2022; 44:295-305. [PMID: 35272550 DOI: 10.1080/08923973.2022.2043896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The role and function of microRNA (miRNA, miR)-140-5p in the calcification of vascular smooth muscle cells (VSMCs) have been explored in this study. METHODS The calcium nodules formed in transfected and β-glycerophosphate (β-GP)-treated VSMCs were observed using Alizarin Red S staining, and alkaline phosphatase (ALP) activity was determined. VSMC apoptosis was detected with flow cytometry assay. The target gene of miR-140-5p was predicted and confirmed with dual-luciferase reporter assay. Relative expressions of miR-140-5p, toll like receptor 4 (TLR4) and vascular calcification-related proteins (α-smooth muscle actin, α-SMA; Msh Homeobox 2, MSX2; bone morphogenetic protein 2, BMP2; Kruppel-like factor 4, KLF4; Runt-related transcription factor 2, RUNX2) were measured through quantitative real time polymerase chain reaction (qRT-PCR) and western blot. RESULTS MiR-140-5p upregulation reversed the effects of β-GP on downregulating miR-140-5p and α-SMA expressions, enhancing ALP activity, calcium nodule formation and cell apoptosis, and upregulating levels of MSX2, BMP2, KLF4 and RUNX2. TLR4 was the target of miR-140-5p, and offset the effects of miR-140-5p on β-GP-induced VSMCs. CONCLUSIONS MiR-140-5p upregulation represses β-GP-induced calcification of VSMCs via targeting TLR4, providing a potential therapeutic method for vascular calcification.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jingxing Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chengxiong Gu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Haibo Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Abdominal Aortic Aneurysm Formation with a Focus on Vascular Smooth Muscle Cells. Life (Basel) 2022; 12:life12020191. [PMID: 35207478 PMCID: PMC8880357 DOI: 10.3390/life12020191] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/29/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a lethal degenerative vascular disease that affects, mostly, the elder population, with a high mortality rate (>80%) upon rupture. It features a dilation of the aortic diameter to larger than 30 mm or more than 50%. Diverse pathological processes are involved in the development of AAA, including aortic wall inflammation, elastin breakdown, oxidative stress, smooth muscle cell (SMC) phenotypic switching and dysfunction, and extracellular matrix degradation. With open surgery being the only therapeutic option up to date, the lack of pharmaceutical treatment approach calls for identifying novel and effective targets and further understanding the pathological process of AAA. Both lifestyle and genetic predisposition have an important role in increasing the risk of AAA. Several cell types are closely related to the pathogenesis of AAA. Among them, vascular SMCs (VSMCs) are gaining much attention as a critical contributor for AAA initiation and/or progression. In this review, we summarize what is known about AAA, including the risk factors, the pathophysiology, and the established animal models of AAA. In particular, we focus on the VSMC phenotypic switching and dysfunction in AAA formation. Further understanding the regulation of VSMC phenotypic changes may provide novel therapeutic targets for the treatment or prevention of AAA.
Collapse
|
16
|
Tanaka D, Ikeda Y, Ikeda E, Yokose M, Ganss B, Iwata T. Effect of Amelotin on Bone Growth in the Murine Calvarial Defect Model. Ann Biomed Eng 2021; 49:3676-3684. [PMID: 34608582 DOI: 10.1007/s10439-021-02867-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 09/23/2021] [Indexed: 11/26/2022]
Abstract
Amelotin (AMTN) is a protein that is expressed during the maturation of dental enamel and has important role in enamel hydroxyapatite mineralization. However, it is not well understood whether AMTN has a strong mineral-promoting ability in bone. In this study, the effect of AMTN on bone healing was investigated using mice calvarial defect model in vivo, and the expression of bone marker genes and cell proliferation were investigated to clarify the role of AMTN in bone mineralization using mouse osteogenic cells (MC3T3-E1) in vitro. Collagen membranes, with or without recombinant human (rh) AMTN, were applied to calvarial defects created on the parietal bones of C57BL/6N mice. Microcomputed tomography and histological observation revealed that the defect largely filled with mineralized tissue by the rhAMTN-containing membrane in eight weeks. Moreover, CD31 positive cells were observed in the newly formed mineralized tissue and around the rhAMTN-containing membrane. In the presence of rhAMTN, the expression of the Spp1 gene in MC3T3-E1 cells significantly increased within ten days in an osteoinductive medium. Moreover, rhAMTN significantly enhanced MC3T3-E1 cell proliferation. These findings indicate that AMTN positively influences bone repair by promoting hydroxyapatite mineralization.
Collapse
Affiliation(s)
- Daiki Tanaka
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
| | - Yuichi Ikeda
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan.
| | - Eri Ikeda
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mako Yokose
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
| | - Bernhard Ganss
- Faculty of Dentistry and Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8549, Japan
| |
Collapse
|
17
|
Dextromethorphan Reduces Oxidative Stress and Inhibits Uremic Artery Calcification. Int J Mol Sci 2021; 22:ijms222212277. [PMID: 34830159 PMCID: PMC8623041 DOI: 10.3390/ijms222212277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/30/2021] [Accepted: 11/09/2021] [Indexed: 12/29/2022] Open
Abstract
Medial vascular calcification has emerged as a key factor contributing to cardiovascular mortality in patients with chronic kidney disease (CKD). Vascular smooth muscle cells (VSMCs) with osteogenic transdifferentiation play a role in vascular calcification. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors reduce reactive oxygen species (ROS) production and calcified-medium-induced calcification of VSMCs. This study investigates the effects of dextromethorphan (DXM), an NADPH oxidase inhibitor, on vascular calcification. We used in vitro and in vivo studies to evaluate the effect of DXM on artery changes in the presence of hyperphosphatemia. The anti-vascular calcification effect of DXM was tested in adenine-fed Wistar rats. High-phosphate medium induced ROS production and calcification of VSMCs. DXM significantly attenuated the increase in ROS production, the decrease in ATP, and mitochondria membrane potential during the calcified-medium-induced VSMC calcification process (p < 0.05). The protective effect of DXM in calcified-medium-induced VSMC calcification was not further increased by NADPH oxidase inhibitors, indicating that NADPH oxidase mediates the effect of DXM. Furthermore, DXM decreased aortic calcification in Wistar rats with CKD. Our results suggest that treatment with DXM can attenuate vascular oxidative stress and ameliorate vascular calcification.
Collapse
|
18
|
Zhang Y, Tang N, Zhou J. Intermedin1‑47 inhibits high phosphate‑induced vascular smooth muscle cell calcification by regulating Wnt/β‑catenin signaling. Mol Med Rep 2021; 24:733. [PMID: 34414455 DOI: 10.3892/mmr.2021.12373] [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: 11/01/2018] [Accepted: 08/12/2019] [Indexed: 11/05/2022] Open
Abstract
Vascular calcification is a major risk factor for cardiovascular disease and accounts for a large proportion of deaths from cardiovascular disease in patients with chronic kidney disease. The high incidence, rapid progression and irreversibility of vascular smooth muscle cell (VSMC) calcification in patients has attracted attention. In the present study, the effect of intermedin1‑47 (IMD1‑47), an important isoform of intermedin, was investigated on the calcification of rat cardiovascular VSMCs induced by high phosphate (HP). To stimulate osteoblast‑like differentiation and calcification in rat VSMCs, 10 mM β‑sodium glycerophosphate was used. The VSMCs were then treated with three doses of IMD1‑47 and the effects of IMD1‑47 on VSMC calcification, on the expression of osteogenic markers [osteoprotegerin, Runt‑related transcription factor 2 (Runx2) and osteopontin (OPN)] and on alkaline phosphatase (ALP) activity were assessed. HP treatment significantly enhanced the cellular calcium content of VSMCs, the expression of osteogenic markers, and ALP activity, while IMD1‑47 significantly reversed these effects in a dose‑dependent manner. The protein expression levels of Wnt1, Wnt3a and active β‑catenin were determined and it was found that IMD1‑47 significantly inhibited their expression. Following β‑catenin silencing, the protein expression levels Runx2 and OPN were increased compared with the IMD1‑47 treatment alone, indicating a role for the Wnt/β‑catenin pathway in the effects of IMD1‑47 on osteogenic markers. The present study suggested that IMD1‑47 inhibited HP‑induced VSMC calcification by regulating the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Geriatrics, Shanghai Fourth Rehabilitation Hospital, Shanghai 200042, P.R. China
| | - Naiwang Tang
- Department of Respiratory, Central Hospital of Xuhui District, Shanghai 200031, P.R. China
| | - Jinjie Zhou
- Department of Cardiology, Central Hospital of Huangpu District, Shanghai 200002, P.R. China
| |
Collapse
|
19
|
Radvar E, Griffanti G, Tsolaki E, Bertazzo S, Nazhat SN, Addison O, Mata A, Shanahan CM, Elsharkawy S. Engineered In vitro Models for Pathological Calcification: Routes Toward Mechanistic Understanding. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Elham Radvar
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| | - Gabriele Griffanti
- Department of Mining and Materials Engineering Faculty of Engineering McGill University Montreal QC H3A 0C5 Canada
| | - Elena Tsolaki
- Department of Medical Physics and Biomedical Engineering University College London London WC1E 6BT UK
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering University College London London WC1E 6BT UK
| | - Showan N. Nazhat
- Department of Mining and Materials Engineering Faculty of Engineering McGill University Montreal QC H3A 0C5 Canada
| | - Owen Addison
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| | - Alvaro Mata
- School of Pharmacy University of Nottingham Nottingham NG7 2RD UK
| | - Catherine M. Shanahan
- BHF Centre of Research Excellence Cardiovascular Division James Black Centre King's College London London SE1 1UL UK
| | - Sherif Elsharkawy
- Centre for Oral, Clinical and Translational Sciences Faculty of Dentistry, Oral and Craniofacial Sciences King's College London London SE1 1UL UK
| |
Collapse
|
20
|
Dong Q, Liang Q, Chen Y, Li J, Lu L, Huang X, Zhou Q. Bibliometric and Visual Analysis of Vascular Calcification Research. Front Pharmacol 2021; 12:690392. [PMID: 34335257 PMCID: PMC8319769 DOI: 10.3389/fphar.2021.690392] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/05/2021] [Indexed: 12/25/2022] Open
Abstract
Background: Extensive studies related to vascular calcification (VC) were conducted in recent years. However, no bibliometric analysis has systematically investigated this topic. Our study aimed to determine the hotspots and frontiers of VC research in the past decade and provide a reference for future scientific research directions and decision-making in the VC field. Methods: VC studies were acquired from the Web of Science Core Collection. Bibliometric and visual analyses were performed using CiteSpace, VOSviewer, and Microsoft Excel software. Results: A total of 8,238 English articles on VC research published in 2011–2020 were obtained. In the past decade, annual publications and citations showed a significant growth trend, especially in 2018–2020. The most productive country, institution, journal and author are the United States, the University of California System, PLOS ONE, and Budoff MJ, respectively. The most frequently cited country, journal, and author are the United States, Journal of the American College of Cardiology, and Floege J, respectively. “Vascular calcification,” “atherosclerosis,” “chronic kidney disease,” and “cardiovascular disease” are the primary keywords. The burst keywords “revascularization,” “calciprotein particle,” “microRNA,” and “microcalcification” are speculated to be the research frontiers. Conclusion: The main research hotspots in the VC field are the molecular mechanisms and prognosis of VC in patients with chronic kidney disease or cardiovascular disease. In addition, endovascular therapy and the development of new drugs targeting signal pathways for VC will become the focus of future research. Moreover, non-coding RNAs related to the diagnosis and treatment of VC are great research prospects.
Collapse
Affiliation(s)
- Qian Dong
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Ying Chen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinhe Li
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Lihe Lu
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xiongqing Huang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qin Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
21
|
Hu CT, Shao YD, Liu YZ, Xiao X, Cheng ZB, Qu SL, Huang L, Zhang C. Oxidative stress in vascular calcification. Clin Chim Acta 2021; 519:101-110. [PMID: 33887264 DOI: 10.1016/j.cca.2021.04.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/28/2022]
Abstract
Vascular calcification (VC), which is closely associated with significant mortality in cardiovascular disease, chronic kidney disease (CKD), and/or diabetes mellitus, is characterized by abnormal deposits of hydroxyapatite minerals in the arterial wall. The impact of oxidative stress (OS) on the onset and progression of VC has not been well described. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, xanthine oxidases, myeloperoxidase (MPO), nitric oxide synthases (NOSs), superoxide dismutase (SOD) and paraoxonases (PONs) are relevant factors that influence the production of reactive oxygen species (ROS). Furthermore, excess ROS-induced OS has emerged as a critical mediator promoting VC through several mechanisms, including phosphate balance, differentiation of vascular smooth muscle cells (VSMCs), inflammation, DNA damage, and extracellular matrix remodeling. Because OS is a significant regulator of VC, antioxidants may be considered as novel treatment options.
Collapse
Affiliation(s)
- Chu-Ting Hu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Medical Laboratory, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yi-Duo Shao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Stomatology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yi-Zhang Liu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Xuan Xiao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Zhe-Bin Cheng
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Stomatology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Lab for Clinical & Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| |
Collapse
|
22
|
Zhou K, Zhu X, Ma K, Liu J, Nürnberg B, Gawaz M, Lang F. Effect of MgCl 2 and GdCl 3 on ORAI1 Expression and Store-Operated Ca 2+ Entry in Megakaryocytes. Int J Mol Sci 2021; 22:ijms22073292. [PMID: 33804889 PMCID: PMC8036595 DOI: 10.3390/ijms22073292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/14/2021] [Accepted: 03/18/2021] [Indexed: 01/07/2023] Open
Abstract
In chronic kidney disease, hyperphosphatemia upregulates the Ca2+ channel ORAI and its activating Ca2+ sensor STIM in megakaryocytes and platelets. ORAI1 and STIM1 accomplish store-operated Ca2+ entry (SOCE) and play a key role in platelet activation. Signaling linking phosphate to upregulation of ORAI1 and STIM1 includes transcription factor NFAT5 and serum and glucocorticoid-inducible kinase SGK1. In vascular smooth muscle cells, the effect of hyperphosphatemia on ORAI1/STIM1 expression and SOCE is suppressed by Mg2+ and the calcium-sensing receptor (CaSR) agonist Gd3+. The present study explored whether sustained exposure to Mg2+ or Gd3+ interferes with the phosphate-induced upregulation of NFAT5, SGK1, ORAI1,2,3, STIM1,2 and SOCE in megakaryocytes. To this end, human megakaryocytic Meg-01 cells were treated with 2 mM ß-glycerophosphate for 24 h in the absence and presence of either 1.5 mM MgCl2 or 50 µM GdCl3. Transcript levels were estimated utilizing q-RT-PCR, protein abundance by Western blotting, cytosolic Ca2+ concentration ([Ca2+]i) by Fura-2 fluorescence and SOCE from the increase in [Ca2+]i following re-addition of extracellular Ca2+ after store depletion with thapsigargin (1 µM). As a result, Mg2+ and Gd3+ upregulated CaSR and blunted or virtually abolished the phosphate-induced upregulation of NFAT5, SGK1, ORAI1,2,3, STIM1,2 and SOCE in megakaryocytes. In conclusion, Mg2+ and the CaSR agonist Gd3+ interfere with phosphate-induced dysregulation of [Ca2+]i in megakaryocytes.
Collapse
Affiliation(s)
- Kuo Zhou
- Department of Pharmacology, Experimental Therapy & Toxicology, Eberhard Karls University, 72074 Tübingen, Germany; (K.Z.); (X.Z.); (K.M.); (B.N.)
| | - Xuexue Zhu
- Department of Pharmacology, Experimental Therapy & Toxicology, Eberhard Karls University, 72074 Tübingen, Germany; (K.Z.); (X.Z.); (K.M.); (B.N.)
| | - Ke Ma
- Department of Pharmacology, Experimental Therapy & Toxicology, Eberhard Karls University, 72074 Tübingen, Germany; (K.Z.); (X.Z.); (K.M.); (B.N.)
| | - Jibin Liu
- Institute of Preventive Veterinary Medicine, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China;
| | - Bernd Nürnberg
- Department of Pharmacology, Experimental Therapy & Toxicology, Eberhard Karls University, 72074 Tübingen, Germany; (K.Z.); (X.Z.); (K.M.); (B.N.)
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany;
| | - Florian Lang
- Department of Vegetative and Clinical Physiology, Eberhard Karls University, 72074 Tübingen, Germany
- Correspondence: ; Tel.: +49-707-129-72194
| |
Collapse
|
23
|
Elashry MI, Gaertner K, Klymiuk MC, Eldaey A, Wenisch S, Arnhold S. Characterisation of stemness and multipotency of ovine muscle-derived stem cells from various muscle sources. J Anat 2021; 239:336-350. [PMID: 33641201 PMCID: PMC8273587 DOI: 10.1111/joa.13420] [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: 08/03/2020] [Revised: 02/16/2021] [Accepted: 02/16/2021] [Indexed: 12/25/2022] Open
Abstract
Muscle stem cells (MSCs) are a promising tool for cell‐based therapy and tissue regeneration in veterinary medicine. Evaluation of MSCs from muscles of different origins improves our understanding of their regenerative potential. The present study compared the stemness, cell proliferation, migration potential, myogenic differentiation (MD), and multipotency of MSCs for four developmentally different muscles of ovine origin. MSCs were isolated from the hind limb (HL), diaphragm (DI), extraocular (EO), and masseter (MS) muscles. Cell proliferation, migration, and stemness were examined using sulforhodamine B, and colony formation assays. Evaluation of multipotency was examined using histological and morphometric analyses, alkaline phosphatase (ALP) activity, and the expression of myogenic, adipogenic, and osteogenic markers using RT‐qPCR. Data were statistically analysed using analysis of variance. The results revealed that all experimental groups expressed stem cell markers paired box transcription factor Pax7, α7‐integrin, CD90, and platelet‐derived growth factor receptor alpha. DI and HL muscle cells displayed higher proliferation, migration, and colony formation capacities compared to the EO and MS muscle cells. HL and DI muscle cells showed increased MD, as indicated by myotube formation and relative expression of MyoD at day 7 and Myogenin at day 14. Although MS and EO muscle cells displayed impaired MD, these cells were more prone to adipogenic differentiation, as indicated by Oil Red O staining and upregulated fatty acid‐binding protein 4 and peroxisome proliferator‐activated receptor gamma expression. DI muscle cells demonstrated a higher osteogenic differentiation capability, as shown by the upregulation of osteopontin expression and an elevated ALP activity. Our data indicate that ovine HL and DI MSCs have a higher regenerative and multipotent potential than the EO and MS muscle cells. These results could be valuable for regional muscle biopsies and cell‐based therapies.
Collapse
Affiliation(s)
- Mohamed I Elashry
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Kateryna Gaertner
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Michele C Klymiuk
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Asmaa Eldaey
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, Germany.,Anatomy and Embryology Department, Faculty of Veterinary Medicine, University of Mansoura, Mansoura, Egypt
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
24
|
Chen JY, Wang YX, Ren KF, Wang YB, Fu GS, Ji J. The influence of substrate stiffness on osteogenesis of vascular smooth muscle cells. Colloids Surf B Biointerfaces 2021; 197:111388. [DOI: 10.1016/j.colsurfb.2020.111388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/29/2020] [Accepted: 09/26/2020] [Indexed: 11/29/2022]
|
25
|
Qiu X, Xu Q, Xu T, Wan P, Sheng Z, Han Y, Yao L. Metformin alleviates β-glycerophosphate-induced calcification of vascular smooth muscle cells via AMPK/mTOR-activated autophagy. Exp Ther Med 2020; 21:58. [PMID: 33365058 PMCID: PMC7716633 DOI: 10.3892/etm.2020.9490] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 09/11/2020] [Indexed: 12/20/2022] Open
Abstract
The aim of the present study was to investigate the effect of metformin on β-glycerophosphate-induced calcification of vascular smooth muscle cells (VSMCs) and the possible mechanisms underlying this. Using an established VSMC calcification model, VSMCs were first treated with β-glycerophosphate, before metformin, 3-methyladenine and compound C were added to the cell cultures in different combinations. Calcium deposition in the cells was examined by Alizarin Red S staining and using the O-cresolphthalein complexone method. To assess the occurrence of autophagy, autophagosomes inside the cells were studied using a transmission electron microscope and green fluorescent microtubule-associated protein 1 light chain 3 (LC3) puncta were examined using a fluorescent microscope. Additionally, protein expression levels of α-smooth muscle actin (α-SMA), runt-related transcription factor 2 (RUNX2), LC3II/I, beclin 1 and 5' adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway-associated proteins were determined by western blot analysis. Metformin increased the number of autophagosomes, green fluorescent LC3 puncta and the levels of LC3II/I, beclin 1, α-SMA and phosphorylated (p)-AMPK in the VSMCs that were treated with β-glycerophosphate when compared to controls; whereas, calcium deposition and the expression levels of RUNX2 and p-mTOR were found to be decreased. Treating the VSMCs with 3-methyladenine or compound C reversed the effects of metformin. The results of the present study suggested that metformin may alleviate β-glycerophosphate-induced calcification of VSMCs, which may be attributed to the activation of AMPK/mTOR signaling pathway-dependent autophagy.
Collapse
Affiliation(s)
- Xiaobo Qiu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China.,Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Qing Xu
- Department of Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Tianhua Xu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Pengzhi Wan
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Zitong Sheng
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Yiran Han
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| |
Collapse
|
26
|
|
27
|
Uremic serum-induced calcification of human aortic smooth muscle cells is a regulated process involving Klotho and RUNX2. Biosci Rep 2020; 39:BSR20190599. [PMID: 31519772 PMCID: PMC6822531 DOI: 10.1042/bsr20190599] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 01/17/2023] Open
Abstract
Vascular calcification (VC) is common in subjects with chronic kidney disease (CKD) and is associated with increased cardiovascular risk. It is an active process involving transdifferentiation of arterial smooth muscle cells (SMCs) into osteogenic phenotype. We investigated the ability of serum from CKD subjects to induce calcification in human SMCs in vitro (calcific potential of sera: CP), and associated changes in expression of Runt-related transcription factor 2 (RUNX2), SM22α, and Klotho. Sera from subjects with CKD (18 stage 3, 17 stage 4/5, and 29 stage 5D) and 20 controls were added to human cultured SMCs and CP quantified. The CP of CKD sera was greater (P<0.01) than that of controls, though not influenced by CKD stage. Modification of diet in renal disease estimated glomerular filtration rate (MDRD-4 eGFR) (P<0.001), serum phosphate (P=0.042), receptor activator of nuclear factor κappa-B ligand (RANKL) (P=0.001), parathyroid hormone (PTH) (P=0.014), and high-density lipoprotein (HDL)/cholesterol ratio (P=0.026) were independent predictors of CP accounting for 45% of variation. Adding calcification buffer (CB: calcium chloride [7 mM] and β-glycerophosphate [7 mM]) increased the CP of control sera to approximate that of CKD sera. CP of CKD sera was unchanged. CKD sera increased RUNX2 expression (P<0.01) in human SMCs and decreased SM22α expression (P<0.05). Co-incubating control but not CKD serum with CB further increased RUNX2 expression (P<0.01). Both SM22α and Klotho expression decreased significantly (P<0.01) in the presence of CKD serum, and were virtually abolished with stage 5D sera. These findings support active regulation by CKD serum of in vitro VC by induction of RUNX2 and suppression of SM22α and Klotho.
Collapse
|
28
|
Cheng CI, Chang HR, Tai MH, Chou MH, Chen GT, Chen PH, Kao YH. Hepatoma-derived growth factor enhances osteoblastic transformation of rat aortic vascular smooth muscle cells in vitro. Life Sci 2020; 256:117964. [DOI: 10.1016/j.lfs.2020.117964] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/02/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023]
|
29
|
Huang C, Zhan JF, Chen YX, Xu CY, Chen Y. LncRNA-SNHG29 inhibits vascular smooth muscle cell calcification by downregulating miR-200b-3p to activate the α-Klotho/FGFR1/FGF23 axis. Cytokine 2020; 136:155243. [PMID: 32818704 DOI: 10.1016/j.cyto.2020.155243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Vascular calcification (VC) is characterized by mineral accumulation on the walls of arteries and veins, which is a pathological process commonly found in elderly individuals and patients with atherosclerosis, hypertension, and diabetes. Emerging evidence suggests that long non-coding RNAs (lncRNAs) play an important role in VC. However, the role of SNHG29 is less clear. METHODS The expression of SNHG29, miR-200b-3p, α-Klotho, FGFR1 and FGF23 in vascular smooth muscle cells (VSMCs) was quantified by qRT-PCR and western blot assays. β-GP was used to construct an in vitro calcification model, followed by MTT assay to detect cell viability. Calcification was determined by alizarin red S staining and quantified by calcification assay. ALP activity was investigated by ALP staining. The interactions among SNHG29, miR-200b-3p and α-Klotho were verified by luciferase assay. RESULTS In the in vitro calcification model, SNHG29 was downregulated, while miR-200b-3p was upregulated. SNHG29 overexpression and miR-200b-3p knockdown significantly suppressed osteoblast-related factors (RUNX2 and BMP2), accompanied by activation of the α-Klotho/FGFR1/FGF23 axis, further inhibiting the formation of calcified nodules. Moreover, miR-200b-3p overexpression and α-Klotho knockdown reversed the SNHG29 overexpression-induced inhibitory effects on calcified VSMCs. CONCLUSION Our study is the first to demonstrate that SNHG29 could inhibit VSMC calcification by downregulating miR-200b-3p to activate the α-Klotho/FGFR1/FGF23 axis, suggesting SNHG29 as a novel therapeutic target for VC-associated diseases.
Collapse
Affiliation(s)
- Chong Huang
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Jin-Feng Zhan
- Physical Examination Center of the Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Yan-Xia Chen
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Cheng-Yun Xu
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Yan Chen
- Department of Nephrology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China.
| |
Collapse
|
30
|
He P, Yu H, Jiang L, Chen Z, Wang S, Macrae VE, Fu X, Zhu D. Hdac9 inhibits medial artery calcification through down-regulation of Osterix. Vascul Pharmacol 2020; 132:106775. [PMID: 32702412 DOI: 10.1016/j.vph.2020.106775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/15/2020] [Accepted: 07/15/2020] [Indexed: 02/03/2023]
Abstract
BACKGROUNDS Medial artery calcification (MAC) significantly contributes to the increased cardiovascular death in patients with chronic kidney disease (CKD). Previous genome-wide association studies have shown that various genetic variants of the histone deacetylase Hdac9 are associated with cardiovascular disease, but the role of Hdac9 in MAC under CKD conditions remains unclear. METHODS High phosphate-induced vascular smooth muscle cell (VSMC) calcification and MAC in mice administered with vitamin D3 (vD) were used in the present study. Alizarin red staining, calcium quantitative assay, qPCR, western blotting and histology were performed. RESULTS Hdac9 expression was significantly down-regulated during high phosphate-induced vascular smooth muscle cell (VSMC) calcification and MAC in mice administered with vitamin D3 (vD). Furthermore, high phosphate treatment inhibited phosphorylation of Akt, and pharmacological inhibition of Akt signaling reduced Hdac9 expression in cultured VSMCs. Knockdown of Hdac9 significantly enhanced calcium deposition in VSMCs. Conversely, adenovirus mediated-overexpression of Hdac9 inhibited high phosphate induced VSMC in vitro calcification. Our subsequent mechanistic studies revealed that the anti-calcific effect of Hdac9 was mediated through down-regulation of osteoblast-specific transcription factor Osterix. CONCLUSION These data suggest that Hdac9 is a novel inhibitor of MAC and may represent a potential therapeutic target for MAC in CKD patients.
Collapse
Affiliation(s)
- Pengcheng He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Hongjiao Yu
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Forester hill, Aberdeen AB25 2ZD, UK
| | - Lei Jiang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Ziying Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Siying Wang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Vicky E Macrae
- The Roslin Institute, RDSVS, Easter Bush Campus, University of Edinburgh, Midlothian EH25 9RG, UK
| | - Xiaodong Fu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China.
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China.
| |
Collapse
|
31
|
Kim JB, Zhao Q, Nguyen T, Pjanic M, Cheng P, Wirka R, Travisano S, Nagao M, Kundu R, Quertermous T. Environment-Sensing Aryl Hydrocarbon Receptor Inhibits the Chondrogenic Fate of Modulated Smooth Muscle Cells in Atherosclerotic Lesions. Circulation 2020; 142:575-590. [PMID: 32441123 DOI: 10.1161/circulationaha.120.045981] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Smooth muscle cells (SMC) play a critical role in atherosclerosis. The Aryl hydrocarbon receptor (AHR) is an environment-sensing transcription factor that contributes to vascular development, and has been implicated in coronary artery disease risk. We hypothesized that AHR can affect atherosclerosis by regulating phenotypic modulation of SMC. METHODS We combined RNA-sequencing, chromatin immunoprecipitation followed by sequencing, assay for transposase-accessible chromatin using sequencing, and in vitro assays in human coronary artery SMCs, with single-cell RNA-sequencing, histology, and RNAscope in an SMC-specific lineage-tracing Ahr knockout mouse model of atherosclerosis to better understand the role of AHR in vascular disease. RESULTS Genomic studies coupled with functional assays in cultured human coronary artery SMCs revealed that AHR modulates the human coronary artery SMC phenotype and suppresses ossification in these cells. Lineage-tracing and activity-tracing studies in the mouse aortic sinus showed that the Ahr pathway is active in modulated SMCs in the atherosclerotic lesion cap. Furthermore, single-cell RNA-sequencing studies of the SMC-specific Ahr knockout mice showed a significant increase in the proportion of modulated SMCs expressing chondrocyte markers such as Col2a1 and Alpl, which localized to the lesion neointima. These cells, which we term "chondromyocytes," were also identified in the neointima of human coronary arteries. In histological analyses, these changes manifested as larger lesion size, increased lineage-traced SMC participation in the lesion, decreased lineage-traced SMCs in the lesion cap, and increased alkaline phosphatase activity in lesions in the Ahr knockout in comparison with wild-type mice. We propose that AHR is likely protective based on these data and inference from human genetic analyses. CONCLUSIONS Overall, we conclude that AHR promotes the maintenance of lesion cap integrity and diminishes the disease-related SMC-to-chondromyocyte transition in atherosclerotic tissues.
Collapse
Affiliation(s)
- Juyong Brian Kim
- Division of Cardiovascular Medicine (J.B.K., Q.Z., T.N., M.P., P.C., R.W., S.T., M.N., R.K., T.Q.), Stanford University School of Medicine, CA.,Cardiovascular Institute (J.B.K., P.C., R.W., T.Q.), Stanford University School of Medicine, CA
| | - Quanyi Zhao
- Division of Cardiovascular Medicine (J.B.K., Q.Z., T.N., M.P., P.C., R.W., S.T., M.N., R.K., T.Q.), Stanford University School of Medicine, CA
| | - Trieu Nguyen
- Division of Cardiovascular Medicine (J.B.K., Q.Z., T.N., M.P., P.C., R.W., S.T., M.N., R.K., T.Q.), Stanford University School of Medicine, CA
| | - Milos Pjanic
- Division of Cardiovascular Medicine (J.B.K., Q.Z., T.N., M.P., P.C., R.W., S.T., M.N., R.K., T.Q.), Stanford University School of Medicine, CA
| | - Paul Cheng
- Division of Cardiovascular Medicine (J.B.K., Q.Z., T.N., M.P., P.C., R.W., S.T., M.N., R.K., T.Q.), Stanford University School of Medicine, CA.,Cardiovascular Institute (J.B.K., P.C., R.W., T.Q.), Stanford University School of Medicine, CA
| | - Robert Wirka
- Division of Cardiovascular Medicine (J.B.K., Q.Z., T.N., M.P., P.C., R.W., S.T., M.N., R.K., T.Q.), Stanford University School of Medicine, CA.,Cardiovascular Institute (J.B.K., P.C., R.W., T.Q.), Stanford University School of Medicine, CA
| | - Stanislao Travisano
- Division of Cardiovascular Medicine (J.B.K., Q.Z., T.N., M.P., P.C., R.W., S.T., M.N., R.K., T.Q.), Stanford University School of Medicine, CA
| | - Manabu Nagao
- Division of Cardiovascular Medicine (J.B.K., Q.Z., T.N., M.P., P.C., R.W., S.T., M.N., R.K., T.Q.), Stanford University School of Medicine, CA
| | - Ramendra Kundu
- Division of Cardiovascular Medicine (J.B.K., Q.Z., T.N., M.P., P.C., R.W., S.T., M.N., R.K., T.Q.), Stanford University School of Medicine, CA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine (J.B.K., Q.Z., T.N., M.P., P.C., R.W., S.T., M.N., R.K., T.Q.), Stanford University School of Medicine, CA.,Cardiovascular Institute (J.B.K., P.C., R.W., T.Q.), Stanford University School of Medicine, CA
| |
Collapse
|
32
|
The protective effects of renin-angiotensin system componts on vascular calcification. J Hum Hypertens 2020; 35:410-418. [PMID: 32398767 DOI: 10.1038/s41371-020-0347-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/19/2020] [Accepted: 04/24/2020] [Indexed: 11/09/2022]
Abstract
Renin-angiotensin system (RAS) has important roles in cardiovascular disease. Angiotensin II (Ang II) and angiotensin-(1-7) (Ang-(1-7)) are major effector peptides of RAS. However, the roles of Ang II type 2 receptor (AT2R) need to be further explored and the roles of Ang-(1-7) are still not very clear on vascular calcification (VC). Therefore, we hypothesized they have effects on preventing VC in vivo and in vitro. VC model is established by inorganic phosphate (IP) cultured with vascular smooth muscle cells (VSMC) for in vitro study and by 5/6 nephrectomy in mice for in vivo study. Increased calcified nodules by Alizarin Red S staining and mRNA expressions of bone morphogenetic protein-2 (BMP-2) and osteocalcin (OCN) by reverse transcription polymerase chain reaction in calcified WT VSMC were significantly inhibited in calcified AT2R overexpression (SmAT2) VSMC or after Ang-(1-7) treatment. After 5/6 nephrectomy, the ratio of positive and total area by Alizarin Red S and von Kossa staining and mRNA expressions of BMP-2 and OCN were significantly increased in ApoE/AT2R knockout mice compared with apolipoprotein E knockout mice, and which were significantly inhibited with Ang-(1-7) administration. Both AT2R and Ang-(1-7) have the effects on preventing VC induced by IP, at least in part through inhibiting BMP-2, OCN expressions, and in which Ang-(1-7) had protective roles mainly through Mas receptor rather than AT2R.
Collapse
|
33
|
Research Models for Studying Vascular Calcification. Int J Mol Sci 2020; 21:ijms21062204. [PMID: 32210002 PMCID: PMC7139511 DOI: 10.3390/ijms21062204] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022] Open
Abstract
Calcification of the vessel wall contributes to high cardiovascular morbidity and mortality. Vascular calcification (VC) is a systemic disease with multifaceted contributing and inhibiting factors in an actively regulated process. The exact underlying mechanisms are not fully elucidated and reliable treatment options are lacking. Due to the complex pathophysiology, various research models exist evaluating different aspects of VC. This review aims to give an overview of the cell and animal models used so far to study the molecular processes of VC. Here, in vitro cell culture models of different origins, ex vivo settings using aortic tissue and various in vivo disease-induced animal models are summarized. They reflect different aspects and depict the (patho)physiologic mechanisms within the VC process.
Collapse
|
34
|
Gayrard N, Muyor K, Notarnicola C, Duranton F, Jover B, Argilés À. Optimisation of cell and ex vivo culture conditions to study vascular calcification. PLoS One 2020; 15:e0230201. [PMID: 32143215 PMCID: PMC7060075 DOI: 10.1371/journal.pone.0230201] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/24/2020] [Indexed: 01/28/2023] Open
Abstract
Medial vascular calcification (MVC) is a highly prevalent disease associated with a high risk of severe, potentially lethal, complications. While animal studies may not systematically be circumvented, in vitro systems have been proven useful to study disease physiopathology. In the context of MVC, the absence of a clinically relevant standardized in vitro method prevents the appropriate comparison and overall interpretation of results originating from different experiments. The aim of our study is to establish in vitro models mimicking in vivo vascular calcification and to select the best methods to unravel the mechanisms involved in MVC. Human aortic smooth muscle cells and rat aortic rings were cultured in different conditions. The influence of fetal calf serum (FCS), alkaline phosphatase, phosphate and calcium concentrations in the medium were evaluated. We identified culture conditions, including the herein reported Aorta Calcifying Medium (ACM), which allowed a reproducible and specific medial calcification of aortic explants. Studying cells and aortic explants cultured, the involvement of bone morphogenetic protein 2 (BMP2) pathway, fibrosis and apoptosis processes in in vitro MVC were demonstrated. Expression of osteoblastic markers was also observed suggesting the occurrence of transdifferentiation of smooth muscle cells to osteoblasts in our models. The use of these models will help researchers in the field of vascular calcification to achieve reproducible results and allow result comparison in a more consistent way.
Collapse
Affiliation(s)
- Nathalie Gayrard
- RD – Néphrologie 2, rue de Mûriers, Montpellier, France
- RD – Néphrologie, EA - 7288 BC2M BatD-RDC UFR - Pharmacie, Montpellier, France
- * E-mail:
| | - Karen Muyor
- RD – Néphrologie 2, rue de Mûriers, Montpellier, France
- RD – Néphrologie, EA - 7288 BC2M BatD-RDC UFR - Pharmacie, Montpellier, France
| | - Cécile Notarnicola
- PhyMedExp (Physiologie et Médecine Expérimentale Cœur Muscles), INSERM-CNRS-Université Montpellier, IURC, Montpellier, France
| | - Flore Duranton
- RD – Néphrologie 2, rue de Mûriers, Montpellier, France
- RD – Néphrologie, EA - 7288 BC2M BatD-RDC UFR - Pharmacie, Montpellier, France
| | - Bernard Jover
- PhyMedExp (Physiologie et Médecine Expérimentale Cœur Muscles), INSERM-CNRS-Université Montpellier, IURC, Montpellier, France
| | - Àngel Argilés
- RD – Néphrologie 2, rue de Mûriers, Montpellier, France
- RD – Néphrologie, EA - 7288 BC2M BatD-RDC UFR - Pharmacie, Montpellier, France
- Néphrologie Dialyse Saint Guilhem (NDSG), CS 40 339, Sete, France
| |
Collapse
|
35
|
Targeting vascular smooth muscle cell dysfunction with xanthine derivative KMUP-3 inhibits abdominal aortic aneurysm in mice. Atherosclerosis 2020; 297:16-24. [PMID: 32059119 DOI: 10.1016/j.atherosclerosis.2020.01.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 01/15/2020] [Accepted: 01/30/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND AND AIMS Inflammation, oxidative stress, matrix degradation, medial calcification and vascular smooth muscle cell (VSMC) loss are prominent features in abdominal aortic aneurysm (AAA). VSMC phenotypic switch to a proinflammatory state and VSMC apoptosis could be targetable mechanisms implicated in the pathogenesis of AAA formation. Herein, we investigated the hypothesis that a xanthine derivative (KMUP-3) might suppress AAA through inhibition of VSMC phenotypic switch and apoptosis. METHODS In vitro, VSMC calcification was induced using β-glycerophosphate. In vivo, AAA was induced using angiotensin II (1000 ng/kg per minute) infusion for 4 weeks in apolipoprotein E-deficient mice. RESULTS As determined by alizarin red S staining and calcium content measurements, KMUP-3 suppressed VSMC calcification. During VSMC calcification, KMUP-3 inhibited mTOR and β-catenin upregulation, essential for VSMC phenotypic switch, while it enhanced AMP-activated protein kinase (AMPK) activation that protects against VSMC phenotypic switch. Moreover, KMUP-3 attenuated VSMC apoptosis with an increased Bcl-2/Bax ratio and reduced activated caspase-3 expression. During AAA formation, treatment with KMUP-3 inhibited phosphorylated mTOR expression and increased phosphorylated AMPK expression in the medial layer. In addition, KMUP-3 treatment suppressed aortic dilatation together with reduction in proinflammatory cytokines and infiltrating macrophages, attenuation of medial VSMC apoptosis and mitigation of reactive oxygen species generation, matrix-degrading proteinase activities, elastin breakdown and vascular calcification. CONCLUSIONS Treatment with KMUP-3 inhibits aneurysm growth possibly through its interference with signaling pathways involved in VSMC phenotypic switch and apoptosis. These findings provide a proof-of-concept validation for VSMC dysfunction as a potential therapeutic target in AAA.
Collapse
|
36
|
Pelzl L, Sahu I, Ma K, Heinzmann D, Bhuyan AAM, Al-Maghout T, Sukkar B, Sharma Y, Marini I, Rigoni F, Artunc F, Cao H, Gutti R, Voelkl J, Pieske B, Gawaz M, Bakchoul T, Lang F. Beta-Glycerophosphate-Induced ORAI1 Expression and Store Operated Ca 2+ Entry in Megakaryocytes. Sci Rep 2020; 10:1728. [PMID: 32015442 PMCID: PMC6997179 DOI: 10.1038/s41598-020-58384-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 01/03/2020] [Indexed: 12/13/2022] Open
Abstract
Impairment of renal phosphate elimination in chronic kidney disease (CKD) leads to enhanced plasma and tissue phosphate concentration, which in turn up-regulates transcription factor NFAT5 and serum & glucocorticoid-inducible kinase SGK1. The kinase upregulates ORAI1, a Ca2+-channel accomplishing store-operated Ca2+-entry (SOCE). ORAI1 is stimulated following intracellular store depletion by Ca2+-sensors STIM1 and/or STIM2. In megakaryocytes and blood platelets SOCE and thus ORAI1 are powerful regulators of activity. The present study explored whether the phosphate-donor ß-glycerophosphate augments NFAT5, ORAI1,2,3 and/or STIM1,2 expressions and thus SOCE in megakaryocytes. Human megakaryocytic Meg01cells were exposed to 2 mM of phosphate-donor ß-glycerophosphate for 24 hours. Platelets were isolated from blood samples of patients with impaired kidney function or control volunteers. Transcript levels were estimated utilizing q-RT-PCR, cytosolic Ca2+-concentration ([Ca2+]i) by Fura-2-fluorescence, and SOCE from increase of [Ca2+]i following re-addition of extracellular Ca2+ after store depletion with thapsigargin (1 µM). NFAT5 and ORAI1 protein abundance was estimated with Western blots. As a result, ß-glycerophosphate increased NFAT5, ORAI1/2/3, STIM1/2 transcript levels, as well as SOCE. Transcript levels of NFAT5, SGK1, ORAI1/2/3, and STIM1/2 as well as NFAT5 and ORAI1 protein abundance were significantly higher in platelets isolated from patients with impaired kidney function than in platelets from control volunteers. In conclusion, phosphate-donor ß-glycerophosphate triggers a signaling cascade of NFAT5/SGK1/ORAI/STIM, thus up-regulating store-operated Ca2+-entry.
Collapse
Affiliation(s)
- Lisann Pelzl
- Transfusion Medicine, Medical Faculty, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | - Itishri Sahu
- Department of Internal Medicine III, Eberhard Karl University Tuebingen, Tuebingen, Germany.,Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Ke Ma
- Department of Internal Medicine III, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | - David Heinzmann
- Department of Internal Medicine III, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | | | - Tamer Al-Maghout
- Department of Internal Medicine III, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | - Basma Sukkar
- Department of Internal Medicine III, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | - Yamini Sharma
- Department of Internal Medicine III, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | - Irene Marini
- Transfusion Medicine, Medical Faculty, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | - Flaviana Rigoni
- Transfusion Medicine, Medical Faculty, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | - Ferruh Artunc
- Department of Internal Medicine IV, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | - Hang Cao
- Department of Internal Medicine III, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | - Ravi Gutti
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Jakob Voelkl
- Institute for Physiology, Johannes Kepler University, Linz, Austria.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Department of Nephrology and Medical Intensive Care, Charité University Medicine, Berlin, Germany.,Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité University Medicine, Berlin, Germany
| | - Burkert Pieske
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, and Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Berlin, Germany.,Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité University Medicine, Berlin, Germany
| | - Meinrad Gawaz
- Department of Internal Medicine III, Eberhard Karl University Tuebingen, Tuebingen, Germany
| | - Tamam Bakchoul
- Transfusion Medicine, Medical Faculty, Eberhard Karl University Tuebingen, Tuebingen, Germany.,Centre for Clinical Transfusion Medicine, University Hospital of Tuebingen, Tuebingen, Germany
| | - Florian Lang
- Department of Vegetative and Clinical Physiology, Eberhard Karl University Tuebingen, Tuebingen, Germany.
| |
Collapse
|
37
|
Rogers MA, Aikawa E. Cardiovascular calcification: artificial intelligence and big data accelerate mechanistic discovery. Nat Rev Cardiol 2020; 16:261-274. [PMID: 30531869 DOI: 10.1038/s41569-018-0123-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiovascular calcification is a health disorder with increasing prevalence and high morbidity and mortality. The only available therapeutic options for calcific vascular and valvular heart disease are invasive transcatheter procedures or surgeries that do not fully address the wide spectrum of these conditions; therefore, an urgent need exists for medical options. Cardiovascular calcification is an active process, which provides a potential opportunity for effective therapeutic targeting. Numerous biological processes are involved in calcific disease, including matrix remodelling, transcriptional regulation, mitochondrial dysfunction, oxidative stress, calcium and phosphate signalling, endoplasmic reticulum stress, lipid and mineral metabolism, autophagy, inflammation, apoptosis, loss of mineralization inhibition, impaired mineral resorption, cellular senescence and extracellular vesicles that act as precursors of microcalcification. Advances in molecular imaging and big data technology, including in multiomics and network medicine, and the integration of these approaches are helping to provide a more comprehensive map of human disease. In this Review, we discuss ectopic calcification processes in the cardiovascular system, with an emphasis on emerging mechanistic knowledge obtained through patient data and advances in imaging methods, experimental models and multiomics-generated big data. We also highlight the potential and challenges of artificial intelligence, machine learning and deep learning to integrate imaging and mechanistic data for drug discovery.
Collapse
Affiliation(s)
- Maximillian A Rogers
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Center for Excellence in Vascular Biology, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Chin DD, Wang J, Mel de Fontenay M, Plotkin A, Magee GA, Chung EJ. Hydroxyapatite-binding micelles for the detection of vascular calcification in atherosclerosis. J Mater Chem B 2019; 7:6449-6457. [PMID: 31553027 PMCID: PMC6812598 DOI: 10.1039/c9tb01918a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Atherosclerosis is a chronic disease characterized by the formation of calcified, arterial plaques. Microcalcifications (5 μm to 100 μm), mainly composed of hydroxyapatite (HA, Ca5(PO4)3(OH)), develop in the fibrous caps of atherosclerotic plaques and can trigger plaque rupture due to the loss of compliance and elasticity. Ultimately, plaque rupture can cause arterial occlusion and embolization and result in ischemic events such as strokes and myocardial infarctions. Unfortunately, current imaging technologies used to detect calcifications are limited by low signal-to-noise ratio or use invasive procedures that pose risk of arterial dissection. To mitigate these drawbacks, in our study, we developed a novel, fluorescently-labeled peptide amphiphile micelle (PAM) that uses a 12 amino acid HA-binding peptide (HABP) [SVSVGMKPSPRP] to target and detect atherosclerotic calcification (HA PAM). Our results show HA PAMs can successfully target HA microcrystals with a strong binding affinity (KD = 6.26 ± 1.2 μM) in vitro. In addition, HA PAMs detected HA mineralization (HA PAM vs. non-targeting micelle, p≤ 0.001; HA PAM vs. scrambled HABP PAM, p≤ 0.01) formed by calcifying mouse aortic vascular smooth muscle cells (MOVAS). Moreover, HA PAMs successfully detected calcifications in atherosclerotic mouse models as well as in patient-derived arteries. Our studies show that HA PAMs show promise as calcium-targeting nanoparticles for the detection of calcifications in atherosclerosis.
Collapse
Affiliation(s)
- Deborah D Chin
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Margot Mel de Fontenay
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Anastasia Plotkin
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gregory A Magee
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA. and Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA and Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA and Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA and Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
39
|
Phadwal K, Feng D, Zhu D, MacRae VE. Autophagy as a novel therapeutic target in vascular calcification. Pharmacol Ther 2019; 206:107430. [PMID: 31647975 DOI: 10.1016/j.pharmthera.2019.107430] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2019] [Indexed: 02/07/2023]
Abstract
The autophagy pathway is a key regulator of cellular metabolism and homeostasis, and plays a critical role in maintaining normal vascular cell function. It is well recognised that autophagy can regulate endothelial cell homeostasis, vascular smooth muscle cell (VSMC) phenotype transition, and calcium (Ca2+) homeostasis in VSMCs. Emerging evidence has demonstrated that autophagy directly protects against vascular calcification (VC). Crosstalk between endosomes, dysfunctional mitochondria, autophagic vesicles and Ca2+ and phosphate (Pi) enriched matrix vesicles (MVs) may underpin the pathogenesis of VC. In this review, we summarize the current experimental evidence in understanding how autophagy maintains normal vascular cell function and its protective role against vascular calcification. We also discuss the underlying molecular and cellular mechanisms through which autophagy inhibits vascular calcification. Pharmacological modulation of autophagy may offer an exciting new strategy for the treatment of vascular calcification.
Collapse
Affiliation(s)
- Kanchan Phadwal
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Du Feng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511436, China.
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Diseases, The Second Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Vicky E MacRae
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| |
Collapse
|
40
|
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
|
41
|
Diverse roles of noncoding RNAs in vascular calcification. Arch Pharm Res 2019; 42:244-251. [PMID: 30673937 DOI: 10.1007/s12272-019-01118-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/15/2019] [Indexed: 12/19/2022]
Abstract
Vascular calcification occurs when calcium phosphate crystals are abnormally deposited in the vessel walls, thus hardening and narrowing the arteries. This condition is commonly observed in patients with diseases such as atherosclerosis, chronic kidney disease, diabetes, and cardiovascular diseases. Despite many studies being conducted, the molecular mechanism involved in vascular calcification is unknown. From recent studies, it is clear that several types of noncoding RNAs are involved in human diseases. It has also been shown that the noncoding RNAs, including microRNAs, long noncoding RNAs, and circular RNAs, are involved in the progression of vascular calcification. With the development of therapeutic approaches based on the manipulation of noncoding RNAs, it is speculated that the modulation of these molecules could be another strategy to treat vascular calcification in the future. In this review, we summarize the roles of various noncoding RNAs in vascular calcification and the technologies to modulate the noncoding RNAs for therapeutic purpose.
Collapse
|
42
|
Enzyme-modified non-oxidized LDL (ELDL) induces human coronary artery smooth muscle cell transformation to a migratory and osteoblast-like phenotype. Sci Rep 2018; 8:11954. [PMID: 30097618 PMCID: PMC6086911 DOI: 10.1038/s41598-018-30073-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/11/2018] [Indexed: 12/29/2022] Open
Abstract
Enzyme modified non-oxidative LDL (ELDL) is effectively taken up by vascular smooth muscle cells (SMC) and mediates transition into foam cells and produces phenotypic changes in SMC function. Our data show that incubation of human coronary artery SMC (HCASMC) with low concentration of ELDL (10 μg/ml) results in significantly enhanced foam cell formation compared to oxidized LDL (200 μg/ml; p < 0.01) or native LDL (200 μg/ml; p < 0.01). Bioinformatic network analysis identified activation of p38 MAPK, NFkB, ERK as top canonical pathways relevant for biological processes linked to cell migration and osteoblastic differentiation in ELDL-treated cells. Functional studies confirmed increased migration of HCASMC upon stimulation with ELDL (10 μg/ml) or Angiopoietin like protein 4, (ANGPTL4, 5 μg/ml), and gain in osteoblastic gene profile with significant increase in mRNA levels for DMP-1, ALPL, RUNX2, OPN/SPP1, osterix/SP7, BMP and reduction in mRNA for MGP and ENPP1. Enhanced calcification of HCASMC by ELDL was demonstrated by Alizarin Red staining. In summary, ELDL is highly potent in inducing foam cells in HCASMC and mediates a phenotypic switch with enhanced migration and osteoblastic gene profile. These results point to the potential of ELDL to induce migratory and osteoblastic effects in human smooth muscle cells with potential implications for migration and calcification of SMCs in human atherosclerosis.
Collapse
|
43
|
Furmanik M, Shanahan CM. ER stress regulates alkaline phosphatase gene expression in vascular smooth muscle cells via an ATF4-dependent mechanism. BMC Res Notes 2018; 11:483. [PMID: 30012221 PMCID: PMC6048897 DOI: 10.1186/s13104-018-3582-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/06/2018] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE Vascular calcification is the deposition of hydroxyapatite crystals in the blood vessel wall. Osteogenic differentiation of vascular smooth muscle cells (VSMCs) plays a key role in this process. Increased expression of alkaline phosphatase (ALP) occurs in some in vitro models of VSMC calcification and is thought to be crucial for mineralization, however, little is known about the transcriptional regulation of ALP in VSMCs. Recently, ALP upregulation was shown to coincide with endoplasmic reticulum (ER) stress-mediated vascular calcification, specifically with expression of the transcription factor ATF4. As no direct links between ALP expression and ER stress have previously been demonstrated in VSMCs, the aim of this study was to investigate whether ATF4 interacts directly with the ALP promoter. RESULTS The present study shows that ALP mRNA and activity were significantly increased by ER stress treatment of human primary VSMCs in vitro and that this was ATF4-dependent. Bioinformatics analysis predicted two ATF4 binding sites in ER-stress responsive regions of the ALP promoter (- 3631 to - 2048 bp from the first intron). However, we found that ATF4 does not bind within this fragment of the ALP promoter region.
Collapse
Affiliation(s)
- Malgorzata Furmanik
- Cardiovascular Division, James Black Centre, King’s College London, 125 Coldharbour Lane, London, SE5 9NU UK
- Present Address: Department of Biochemistry, CARIM-Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Catherine M. Shanahan
- Cardiovascular Division, James Black Centre, King’s College London, 125 Coldharbour Lane, London, SE5 9NU UK
| |
Collapse
|
44
|
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
|
45
|
Wang J, Zhou JJ, Robertson GR, Lee VW. Vitamin D in Vascular Calcification: A Double-Edged Sword? Nutrients 2018; 10:nu10050652. [PMID: 29786640 PMCID: PMC5986531 DOI: 10.3390/nu10050652] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 01/13/2023] Open
Abstract
Vascular calcification (VC) as a manifestation of perturbed mineral balance, is associated with aging, diabetes and kidney dysfunction, as well as poorer patient outcomes. Due to the current limited understanding of the pathophysiology of vascular calcification, the development of effective preventative and therapeutic strategies remains a significant clinical challenge. Recent evidence suggests that traditional risk factors for cardiovascular disease, such as left ventricular hypertrophy and dyslipidaemia, fail to account for clinical observations of vascular calcification. Therefore, more complex underlying processes involving physiochemical changes to mineral balance, vascular remodelling and perturbed hormonal responses such as parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF-23) are likely to contribute to VC. In particular, VC resulting from modifications to calcium, phosphate and vitamin D homeostasis has been recently elucidated. Notably, deregulation of vitamin D metabolism, dietary calcium intake and renal mineral handling are associated with imbalances in systemic calcium and phosphate levels and endothelial cell dysfunction, which can modulate both bone and soft tissue calcification. This review addresses the current understanding of VC pathophysiology, with a focus on the pathogenic role of vitamin D that has provided new insights into the mechanisms of VC.
Collapse
Affiliation(s)
- Jeffrey Wang
- Centre for Transplantation and Renal Research, Westmead Institute of Medical Research, Westmead, NSW 2145, Australia.
| | - Jimmy J Zhou
- Centre for Transplantation and Renal Research, Westmead Institute of Medical Research, Westmead, NSW 2145, Australia.
- Centre for Kidney Research, Children's Hospital at Westmead, Westmead, NSW 2145, Australia.
| | | | - Vincent W Lee
- Centre for Transplantation and Renal Research, Westmead Institute of Medical Research, Westmead, NSW 2145, Australia.
| |
Collapse
|
46
|
A novel bioactive osteogenesis scaffold delivers ascorbic acid, β-glycerophosphate, and dexamethasone in vivo to promote bone regeneration. Oncotarget 2018; 8:31612-31625. [PMID: 28404942 PMCID: PMC5458234 DOI: 10.18632/oncotarget.15779] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 01/17/2017] [Indexed: 12/28/2022] Open
Abstract
Ascorbic acid, β-glycerophosphate, and dexamethasone have been used in osteogenesis differentiation medium for in vitro cell culture, nothing is known for delivering these three bioactive compounds in vivo. In this study, we synthesized a novel bioactive scaffold by combining these three compounds with a lysine diisocyanate-based polyurethane. These bioactive compounds were released from the scaffold during the degradation process. The cell culture showed that the sponge-like structure in the scaffold was critical in providing a large surface area to support cell growth and all degradation products of the polymer were non-toxic. This bioactive scaffold enhanced the bone regeneration as evidenced by increasing the expression of three bone-related genes including collagen type I, Runx-2 and osteocalcin in rabbit bone marrow stem cells (BMSCs) in vitro and in vivo. The osteogenesis differentiation of BMSCs cultured in this bioactive scaffold was similar to that in osteogenesis differentiation medium and more extensive in this bioactive scaffold compared to the scaffold without these three bioactive compounds. These results indicated that the scaffold containing three bioactive compounds was a good osteogenesis differentiation promoter to enhance the osteogenesis differentiation and new bone formation in vivo.
Collapse
|
47
|
Wen J, Zhao Z, Huang L, Li L, Li J, Zeng Y, Wu J, Miao Y. Switch of the ovarian cancer cell to a calcifying phenotype in the calcification of ovarian cancer. J Cancer 2018; 9:1006-1016. [PMID: 29581780 PMCID: PMC5868168 DOI: 10.7150/jca.22932] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/28/2018] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE The main aim of this study was to study swith of the ovarian cancer cell to a calcifying phenotype in the formation of calcification in ovarian cancer, and to offer some help for ovarian cancer's diagnosis and differentiation therapy. METHODS The mineralization of ovarian cancer cell lines SKOV3 was induced via calcification medium for 21 d in vitro. Alizarin red staining, von kossa staining, calcein fluorescence staining and ALP activity detection were used to identify mineralization in calcification model of ovarian cancer. Also, the changes of ultrastructure and the mineralization biomarkers after the induction of calcification medium were investigated by transmission electron microscopy and western blot, respectively. The SKOV3 cells migration behavior after the induction of calcification medium was evaluated by using transwell assay and scratch wound. Finally, mineralization biomarkers were verified in 40 cases of calcified ovarian cancer specimens and matched 40 non-calcified ovarian cancer tissues. RESULTS Classical calcium salt detection methods confirmed that the culture of SKOV3 cells in calcification medium was an appropriate ovarian cancer calcification model in vitro. Transmission electron microscopy and western blot revealed respectively the presence of cells with morphological characteristics of osteoblasts and the upregulation of mineralization biomarkers expression in treatment group. Transwell assay and scratch wound showed the decreased SKOV3 cell migration in treatment group. In specimens, the calcification occurred predominantly in well-differentiated carcinomas and the expression of the BMP2 and OPN elevated in calcified group. CONCLUSION Our study showed that the switch of the ovarian cancer cell to a calcifying phenotype in the formation of calcification in ovarian cancer. The calcified phenotypic transformation may inform the new prospective in ovarian cancer therapy.
Collapse
Affiliation(s)
- Jirui Wen
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, P.R. China.,West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhiwei Zhao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Liwei Huang
- West China School of Stomatology Medicine, Sichuan University, Chengdu, China
| | - Liang Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jiman Li
- Pathology Department, Sichuan Cancer Hospital, Chengdu, China
| | - Ye Zeng
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jiang Wu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yali Miao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
48
|
Ter Braake AD, Tinnemans PT, Shanahan CM, Hoenderop JGJ, de Baaij JHF. Magnesium prevents vascular calcification in vitro by inhibition of hydroxyapatite crystal formation. Sci Rep 2018; 8:2069. [PMID: 29391410 PMCID: PMC5794996 DOI: 10.1038/s41598-018-20241-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/15/2018] [Indexed: 01/01/2023] Open
Abstract
Magnesium has been shown to effectively prevent vascular calcification associated with chronic kidney disease. Magnesium has been hypothesized to prevent the upregulation of osteoblastic genes that potentially drives calcification. However, extracellular effects of magnesium on hydroxyapatite formation are largely neglected. This study investigated the effects of magnesium on intracellular changes associated with transdifferentiation and extracellular crystal formation. Bovine vascular smooth muscle cells were calcified using β-glycerophosphate. Transcriptional analysis, alkaline phosphatase activity and detection of apoptosis were used to identify transdifferentiation. Using X-ray diffraction and energy dispersive spectroscopy extracellular crystal composition was investigated. Magnesium prevented calcification in vascular smooth muscle cells. β-glycerophosphate increased expression of osteopontin but no other genes related to calcification. Alkaline phosphatase activity was stable and apoptosis was only detected after calcification independent of magnesium. Blocking of the magnesium channel TRPM7 using 2-APB did not abrogate the protective effects of magnesium. Magnesium prevented the formation of hydroxyapatite, which formed extensively during β-glycerophosphate treatment. Magnesium reduced calcium and phosphate fractions of 68% and 41% extracellular crystals, respectively, without affecting the fraction of magnesium. This study demonstrates that magnesium inhibits hydroxyapatite formation in the extracellular space, thereby preventing calcification of vascular smooth muscle cells.
Collapse
Affiliation(s)
- Anique D Ter Braake
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Paul T Tinnemans
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Catherine M Shanahan
- BHF Centre of Research Excellence, Cardiovascular Division, James Black Centre, King's College, London, United Kingdom
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
49
|
Kamenskiy A, Poulson W, Sim S, Reilly A, Luo J, MacTaggart J. Prevalence of Calcification in Human Femoropopliteal Arteries and its Association with Demographics, Risk Factors, and Arterial Stiffness. Arterioscler Thromb Vasc Biol 2018; 38:e48-e57. [PMID: 29371245 DOI: 10.1161/atvbaha.117.310490] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/15/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Arterial calcification and stiffening increase the risk of reconstruction failure, amputation, and mortality in patients with peripheral arterial disease, but underlying mechanisms and prevalence are unclear. APPROACH AND RESULTS Fresh human femoropopliteal arteries were obtained from n=431 tissue donors aged 13 to 82 years (mean age, 53±16 years) recording the in situ longitudinal prestretch. Arterial diameter, wall thickness, and opening angles were measured optically, and stiffness was assessed using planar biaxial extension and constitutive modeling. Histological features were determined using transverse and longitudinal Verhoeff-Van Gieson and Alizarin stains. Medial calcification was quantified using a 7-stage grading scale and was correlated with structural and mechanical properties and clinical characteristics. Almost half (46%) of the femoropopliteal arteries had identifiable medial calcification. Older arteries were more calcified, but small calcium deposits were observed in arteries as young as 18 years old. After controlling for age, positive correlations were observed between calcification, diabetes mellitus, dyslipidemia, and body mass index. Tobacco use demonstrated a negative correlation. Calcified arteries were larger in diameter but had smaller circumferential opening angles. They were also stiffer longitudinally and circumferentially and had thinner tunica media and external elastic lamina with more discontinuous elastic fibers. CONCLUSIONS Although aging is the dominant risk factor for femoropopliteal artery calcification and stiffening, these processes seem to be linked and can begin at a young age. Calcification is associated with the presence of certain risk factors and with elastic fiber degradation, suggesting overlapping molecular pathways that require further investigation.
Collapse
Affiliation(s)
- Alexey Kamenskiy
- From the Department of Surgery, University of Nebraska Medical Center, Omaha.
| | - William Poulson
- From the Department of Surgery, University of Nebraska Medical Center, Omaha
| | - Sylvie Sim
- From the Department of Surgery, University of Nebraska Medical Center, Omaha
| | - Austin Reilly
- From the Department of Surgery, University of Nebraska Medical Center, Omaha
| | - Jiangtao Luo
- From the Department of Surgery, University of Nebraska Medical Center, Omaha
| | - Jason MacTaggart
- From the Department of Surgery, University of Nebraska Medical Center, Omaha.
| |
Collapse
|
50
|
Salimi F, Jafari-Nodooshan S, Zohourian N, Kolivand S, Hamedi J. Simultaneous anti-diabetic and anti-vascular calcification activity of Nocardia sp. UTMC 751. Lett Appl Microbiol 2018; 66:110-117. [PMID: 29223135 DOI: 10.1111/lam.12833] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/04/2017] [Accepted: 11/15/2017] [Indexed: 02/03/2023]
Abstract
Alpha-amylase can act as a significant player in causing hyperglycaemia, leading to protein glycation, which is the main complication in this condition, besides causing vascular calcification (VC), an important vascular failure caused due to this. In order to find a natural source of the biocompounds with inhibitory effects on α-amylase, 15 fermentation broth extracts of actinobacteria (FBEA) (200 μg ml-1 ) have been screened. Finally, the effects of the most efficient FBE have been investigated on osteopontin (OPN, a VC marker) mRNA level in the vascular smooth muscle cells under the calcification conditions, and the chemical constituents of the most efficient FBE were analysed using gas chromatography and mass spectrometry (GC-MS) analysis. The tested FBEA showed anti-amylase (7·2-21%) and anti-denaturation (7·5-37%) activities. Among the tested FBEA, Nocardia sp. UTMC 751 FBE showed the highest anti-amylase activity (21%). This treatment group also displayed the minimum fructosamine and the maximum thiol groups content. In addition, this FBE reduced the mRNA level of the OPN (fourfold). The GC-MS analysis demonstrated the existence of three volatile and known antioxidants including pyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-3-(2-methylpropyl)-, pyrrolo[1,2-a]pyrazine-1,4-dione, hexahydro-3-(phenylmethyl)- and methyl ester of 3-(3,5-di-tert-butyl-4-hydroxyphenyl)-propionic acid in the FBE of Nocardia sp. UTMC 751. The results indicated that Nocardia sp. UTMC 751 is a considerable source of bioactive compounds that are effective against the direct and indirect pathological targets involved in diabetes. This study highlights the significant potential of rare Actinomycetes in producing pharmaceutically important biocompounds. SIGNIFICANCE AND IMPACT OF THE STUDY Actinobacteria are one of the best natural libraries for discovering drugs. Various commercial drugs have been developed against infectious and metabolic disorders from actinobacteria; however, there is no report on their simultaneous inhibitory effect against diabetes, a life-threatening disease, and its related pathological processes, like inflammation and vascular calcification (VC). In this research, after several screening, Nocardia sp. UTMC 751 was introduced as the first microbial source exhibiting a simultaneous inhibitory activity on the targets, including hyperglycaemia and protein glycation, and other involved pathological processes like inflammation and VC.
Collapse
Affiliation(s)
- F Salimi
- Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran.,Microbial Technology and Products Research Center, University of Tehran, Tehran, Iran
| | - S Jafari-Nodooshan
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - N Zohourian
- Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran.,Microbial Technology and Products Research Center, University of Tehran, Tehran, Iran
| | - S Kolivand
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - J Hamedi
- Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran.,Microbial Technology and Products Research Center, University of Tehran, Tehran, Iran
| |
Collapse
|