1
|
Liao YR, Tsai YC, Hsieh TH, Tsai MT, Lin FY, Lin SJ, Lin CC, Chiang HY, Chu PH, Li SY. FHL2 in arterial medial calcification in chronic kidney disease. Nephrol Dial Transplant 2024; 39:2025-2039. [PMID: 38664060 PMCID: PMC11596093 DOI: 10.1093/ndt/gfae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Arterial medial calcification (AMC) is a common complication in individuals with chronic kidney disease (CKD), which can lead to cardiovascular morbidity and mortality. The progression of AMC is controlled by a key transcription factor called runt-related transcription factor 2 (RUNX2), which induces vascular smooth muscle cells (VSMCs) transdifferentiation into an osteogenic phenotype. However, RUNX2 has not been targeted for therapy due to its essential role in bone development. The objective of our study was to discover a RUNX2 coactivator that is highly expressed in arterial VSMCs as a potential therapy for AMC. METHODS We employed transcriptomic analysis of human data and an animal reporter system to pinpoint four and a half LIM domains 2 (FHL2) as a potential target. Subsequently, we investigated the mRNA and protein expression patterns of FHL2 in the aortas of both human and animal subjects with CKD. To examine the role of FHL2 in the RUNX2 transcription machinery, we conducted coimmunoprecipitation and chromatin immunoprecipitation experiments. Next, we manipulated FHL2 expression in cultured VSMCs to examine its impact on high phosphate-induced transdifferentiation. Finally, we employed FHL2-null mice to confirm the role of FHL2 in the development of AMC in vivo. RESULTS Among all the potential RUNX2 cofactors, FHL2 displays selective expression within the cardiovascular system. In the context of CKD subjects, FHL2 undergoes upregulation and translocation from the cytosol to the nucleus of arterial VSMCs. Once in the nucleus, FHL2 interacts structurally and functionally with RUNX2, acting as a coactivator of RUNX2. Notably, the inhibition of FHL2 expression averts transdifferentiation of VSMCs into an osteogenic phenotype and mitigates aortic calcification in uremic animals, without causing any detrimental effects on the skeletal system. CONCLUSION These observations provide evidence that FHL2 is a promising target for treating arterial calcification in patients with CKD.
Collapse
MESH Headings
- Animals
- LIM-Homeodomain Proteins/metabolism
- LIM-Homeodomain Proteins/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/complications
- Humans
- Mice
- Muscle Proteins/metabolism
- Muscle Proteins/genetics
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Transcription Factors/metabolism
- Transcription Factors/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cells, Cultured
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/etiology
- Vascular Calcification/genetics
- Male
- Cell Transdifferentiation
- Mice, Inbred C57BL
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Knockout
Collapse
Affiliation(s)
- Yuan-Ru Liao
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Cheng Tsai
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tsung-Han Hsieh
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Ming-Tsun Tsai
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Feng-Yen Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ching Lin
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hou-Yu Chiang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Science, College of Medicine, Chang Guang University, Taoyuan, Taiwan
| | - Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taiwan
| | - Szu-Yuan Li
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
2
|
Sarkar A, Pawar SV, Chopra K, Jain M. Gamut of glycolytic enzymes in vascular smooth muscle cell proliferation: Implications for vascular proliferative diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167021. [PMID: 38216067 DOI: 10.1016/j.bbadis.2024.167021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the media of the blood vessels and are responsible for maintaining vascular tone. Emerging evidence confirms that VSMCs possess high plasticity. During vascular injury, VSMCs switch from a "contractile" phenotype to an extremely proliferative "synthetic" phenotype. The balance between both strongly affects the progression of vascular remodeling in many cardiovascular pathologies such as restenosis, atherosclerosis and aortic aneurism. Proliferating cells demand high energy requirements and to meet this necessity, alteration in cellular bioenergetics seems to be essential. Glycolysis, fatty acid metabolism, and amino acid metabolism act as a fuel for VSMC proliferation. Metabolic reprogramming of VSMCs is dynamically variable that involves multiple mechanisms and encompasses the coordination of various signaling molecules, proteins, and enzymes. Here, we systemically reviewed the metabolic changes together with the possible treatments that are still under investigation underlying VSMC plasticity which provides a promising direction for the treatment of diseases associated with VSMC proliferation. A better understanding of the interaction between metabolism with associated signaling may uncover additional targets for better therapeutic strategies in vascular disorders.
Collapse
Affiliation(s)
- Ankan Sarkar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Sandip V Pawar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Kanwaljit Chopra
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Manish Jain
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| |
Collapse
|
3
|
Wang S, Hu S, Pan Y. The emerging roles of irisin in vascular calcification. Front Endocrinol (Lausanne) 2024; 15:1337995. [PMID: 38405155 PMCID: PMC10884194 DOI: 10.3389/fendo.2024.1337995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/19/2024] [Indexed: 02/27/2024] Open
Abstract
Vascular calcification is a common accompanying pathological change in many chronic diseases, which is caused by calcium deposition in the blood vessel wall and leads to abnormal blood vessel function. With the progress of medical technology, the diagnosis rate of vascular calcification has explosively increased. However, due to its mechanism's complexity, no effective drug can relieve or even reverse vascular calcification. Irisin is a myogenic cytokine regulating adipose tissue browning, energy metabolism, glucose metabolism, and other physiological processes. Previous studies have shown that irisin could serve as a predictor for vascular calcification, and protect against hypertension, diabetes, chronic kidney disease, and other risk factors for vascular calcification. In terms of mechanism, it improves vascular endothelial dysfunction and phenotypic transformation of vascular smooth muscle cells. All the above evidence suggests that irisin plays a predictive and protective role in vascular calcification. In this review, we summarize the association of irisin to the related risk factors for vascular calcification and mainly explore the role of irisin in vascular calcification.
Collapse
Affiliation(s)
- Shuangshuang Wang
- Department of Cardiology, The First People’s Hospital of Wenling (The Affiliated Wenling Hospital of Wenzhou Medical University), Wenling, Zhejiang, China
| | - Siwang Hu
- The Orthopaedic Center, The First People’s Hospital of Wenling (The Affiliated Wenling Hospital of Wenzhou Medical University), Wenling, Zhejiang, China
| | - Yuping Pan
- Department of Internal Medicine, Yuhuan Second People’s Hospital, Yuhuan, Zhejiang, China
| |
Collapse
|
4
|
Urbain F, Ponnaiah M, Ichou F, Lhomme M, Materne C, Galier S, Haroche J, Frisdal E, Mathian A, Durand H, Pha M, Hie M, Kontush A, Cluzel P, Lesnik P, Amoura Z, Guerin M, Cohen Aubart F, Le Goff W. Impaired metabolism predicts coronary artery calcification in women with systemic lupus erythematosus. EBioMedicine 2023; 96:104802. [PMID: 37725854 PMCID: PMC10518349 DOI: 10.1016/j.ebiom.2023.104802] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/23/2023] [Accepted: 09/03/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Patients with systemic lupus erythematosus (SLE) exhibit a high risk for cardiovascular diseases (CVD) which is not fully explained by the classical Framingham risk factors. SLE is characterized by major metabolic alterations which can contribute to the elevated prevalence of CVD. METHODS A comprehensive analysis of the circulating metabolome and lipidome was conducted in a large cohort of 211 women with SLE who underwent a multi-detector computed tomography scan for quantification of coronary artery calcium (CAC), a robust predictor of coronary heart disease (CHD). FINDINGS Beyond traditional risk factors, including age and hypertension, disease activity and duration were independent risk factors for developing CAC in women with SLE. The presence of coronary calcium was associated with major alterations of circulating lipidome dominated by an elevated abundance of ceramides with very long chain fatty acids. Alterations in multiple metabolic pathways, including purine, arginine and proline metabolism, and microbiota-derived metabolites, were also associated with CAC in women with SLE. Logistic regression with bootstrapping of lipidomic and metabolomic variables were used to develop prognostic scores. Strikingly, combining metabolic and lipidomic variables with clinical and biological parameters markedly improved the prediction (area under the curve: 0.887, p < 0.001) of the presence of coronary calcium in women with SLE. INTERPRETATION The present study uncovers the contribution of disturbed metabolism to the presence of coronary artery calcium and the associated risk of CHD in SLE. Identification of novel lipid and metabolite biomarkers may help stratifying patients for reducing CVD morbidity and mortality in SLE. FUNDING INSERM and Sorbonne Université.
Collapse
Affiliation(s)
- Fanny Urbain
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France
| | - Maharajah Ponnaiah
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), ICAN I/O Data Science (MPo), ICAN Omics (FI and ML), 75013, Paris, France
| | - Farid Ichou
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), ICAN I/O Data Science (MPo), ICAN Omics (FI and ML), 75013, Paris, France
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), ICAN I/O Data Science (MPo), ICAN Omics (FI and ML), 75013, Paris, France
| | - Clément Materne
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Sophie Galier
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Julien Haroche
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France
| | - Eric Frisdal
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Alexis Mathian
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France
| | - Herve Durand
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Micheline Pha
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France
| | - Miguel Hie
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France
| | - Anatol Kontush
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Philippe Cluzel
- Cardiovascular and Interventional Radiology Department, Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, F-75013, France
| | - Philippe Lesnik
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Zahir Amoura
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France; Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France
| | - Maryse Guerin
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France
| | - Fleur Cohen Aubart
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Centre de Référence pour le Lupus, le Syndrome des Anti-phospholipides et Autres Maladies Auto-immunes Rares, Service de Médecine Interne 2, Paris, France.
| | - Wilfried Le Goff
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (IHU ICAN), UMR_S1166, F-75013, Paris, France.
| |
Collapse
|
5
|
Chen J, Yu H, Tan X, Mok SWF, Xie Y, Wang Y, Jiang X, Macrae VE, Lan L, Fu X, Zhu D. PFKFB3-driven vascular smooth muscle cell glycolysis promotes vascular calcification via the altered FoxO3 and lactate production. FASEB J 2023; 37:e23182. [PMID: 37682013 DOI: 10.1096/fj.202300900r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/08/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023]
Abstract
A link between increased glycolysis and vascular calcification has recently been reported, but it remains unclear how increased glycolysis contributes to vascular calcification. We therefore investigated the role of PFKFB3, a critical enzyme of glycolysis, in vascular calcification. We found that PFKFB3 expression was upregulated in calcified mouse VSMCs and arteries. We showed that expression of miR-26a-5p and miR-26b-5p in calcified mouse arteries was significantly decreased, and a negative correlation between Pfkfb3 mRNA expression and miR-26a-5p or miR-26b-5p was seen in these samples. Overexpression of miR-26a/b-5p significantly inhibited PFKFB3 expression in VSMCs. Intriguingly, pharmacological inhibition of PFKFB3 using PFK15 or knockdown of PFKFB3 ameliorated vascular calcification in vD3 -overloaded mice in vivo or attenuated high phosphate (Pi)-induced VSMC calcification in vitro. Consistently, knockdown of PFKFB3 significantly reduced glycolysis and osteogenic transdifferentiation of VSMCs, whereas overexpression of PFKFB3 in VSMCs induced the opposite effects. RNA-seq analysis and subsequent experiments revealed that silencing of PFKFB3 inhibited FoxO3 expression in VSMCs. Silencing of FoxO3 phenocopied the effects of PFKFB3 depletion on Ocn and Opg expression but not Alpl in VSMCs. Pyruvate or lactate supplementation, the product of glycolysis, reversed the PFKFB3 depletion-mediated effects on ALP activity and OPG protein expression in VSMCs. Our results reveal that blockade of PFKFB3-mediated glycolysis inhibits vascular calcification in vitro and in vivo. Mechanistically, we show that FoxO3 and lactate production are involved in PFKFB3-driven osteogenic transdifferentiation of VSMCs. PFKFB3 may be a promising therapeutic target for the treatment of vascular calcification.
Collapse
Affiliation(s)
- Jiaxin Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Hongjiao Yu
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, China
| | - Xiao Tan
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Simon Wing Fai Mok
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Yuchen Xie
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yueheng Wang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xueyan Jiang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Vicky E Macrae
- Functional Genetics and Development, The Royal (Dick) School of Veterinary Studies and The Roslin Institute, University of Edinburgh, Midlothian, UK
| | - Lan Lan
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaodong Fu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Jud P, Meinitzer A, Strohmaier H, Arefnia B, Wimmer G, Obermayer-Pietsch B, Foris V, Kovacs G, Odler B, Moazedi-Fürst F, Brodmann M, Hafner F. Association of amino acids and parameters of bone metabolism with endothelial dysfunction and vasculopathic changes in limited systemic sclerosis. Front Med (Lausanne) 2023; 10:1193121. [PMID: 37425312 PMCID: PMC10327605 DOI: 10.3389/fmed.2023.1193121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023] Open
Abstract
Objectives Pathways contributing to endothelial dysfunction in patients with limited cutaneous systemic sclerosis (lcSSc) are largely unknown. The aim of this study was to investigate potential associations of amino acids and parameters of bone metabolism with endothelial dysfunction and vasculopathy-related changes in patients with lcSSc and early-stage vasculopathy. Methods Amino acids, calciotropic parameters, including 25-hydroxyvitamin D and parathyroid hormone (PTH), and bone turnover parameters, including osteocalcin and N-terminal peptide of procollagen-3 (P3NP), were measured in 38 lcSSc patients and 38 controls. Endothelial dysfunction was assessed by biochemical parameters, pulse-wave analysis, flow-mediated and nitroglycerine-mediated dilation. Additionally, vasculopathy-related and SSc-specific clinical changes including capillaroscopic, skin, renal, pulmonary, gastrointestinal and periodontal parameters were recorded. Results No significant differences in amino acids, calciotropic and bone turnover parameters were observed between lcSSc patients and controls. In patients with lcSSc, several significant correlations were found between selected amino acids, parameters of endothelial dysfunction, vasculopathy-related and SSc-specific clinical changes (all with p < 0.05). In addition, significant correlations were observed between PTH and 25-hydroxyvitamin D with homoarginine, and between osteocalcin, PTH and P3NP with modified Rodnan skin score and selected periodontal parameters (all with p < 0.05). Vitamin D deficiency defined as 25-hydroxyvitamin D < 20 ng/ml was associated with the presence of puffy finger (p = 0.046) and early pattern (p = 0.040). Conclusion Selected amino acids may affect endothelial function and may be associated to vasculopathy-related and clinical changes in lcSSc patients, while the association with parameters of bone metabolism seems to be minor.
Collapse
Affiliation(s)
- Philipp Jud
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Heimo Strohmaier
- Center of Medical Research (ZMF), Medical University of Graz, Graz, Austria
| | - Behrouz Arefnia
- Division of Restorative Dentistry, Endodontics, Periodontology and Prosthodontics, Department of Dental Medicine and Oral Health, Medical University of Graz, Graz, Austria
| | - Gernot Wimmer
- Division of Restorative Dentistry, Endodontics, Periodontology and Prosthodontics, Department of Dental Medicine and Oral Health, Medical University of Graz, Graz, Austria
| | - Barbara Obermayer-Pietsch
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Endocrinology Lab Platform, Medical University of Graz, Graz, Austria
| | - Vasile Foris
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Gabor Kovacs
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Balazs Odler
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florentine Moazedi-Fürst
- Division of Rheumatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Marianne Brodmann
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Franz Hafner
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
7
|
Ma J, Li Y, Yang X, Liu K, Zhang X, Zuo X, Ye R, Wang Z, Shi R, Meng Q, Chen X. Signaling pathways in vascular function and hypertension: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:168. [PMID: 37080965 PMCID: PMC10119183 DOI: 10.1038/s41392-023-01430-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension is a global public health issue and the leading cause of premature death in humans. Despite more than a century of research, hypertension remains difficult to cure due to its complex mechanisms involving multiple interactive factors and our limited understanding of it. Hypertension is a condition that is named after its clinical features. Vascular function is a factor that affects blood pressure directly, and it is a main strategy for clinically controlling BP to regulate constriction/relaxation function of blood vessels. Vascular elasticity, caliber, and reactivity are all characteristic indicators reflecting vascular function. Blood vessels are composed of three distinct layers, out of which the endothelial cells in intima and the smooth muscle cells in media are the main performers of vascular function. The alterations in signaling pathways in these cells are the key molecular mechanisms underlying vascular dysfunction and hypertension development. In this manuscript, we will comprehensively review the signaling pathways involved in vascular function regulation and hypertension progression, including calcium pathway, NO-NOsGC-cGMP pathway, various vascular remodeling pathways and some important upstream pathways such as renin-angiotensin-aldosterone system, oxidative stress-related signaling pathway, immunity/inflammation pathway, etc. Meanwhile, we will also summarize the treatment methods of hypertension that targets vascular function regulation and discuss the possibility of these signaling pathways being applied to clinical work.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanan Li
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiangyu Yang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin Zhang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xianghao Zuo
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Runyu Ye
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ziqiong Wang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Rufeng Shi
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qingtao Meng
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
8
|
Koch V, Gruenewald LD, Gruber-Rouh T, Eichler K, Leistner DM, Mahmoudi S, Booz C, Bernatz S, D'Angelo T, Albrecht MH, Alizadeh LS, Nour-Eldin NEA, Scholtz JE, Yel I, Vogl TJ, März W, Hardt SE, Martin SS. Homoarginine in the cardiovascular system: Pathophysiology and recent developments. Fundam Clin Pharmacol 2022; 37:519-529. [PMID: 36509694 DOI: 10.1111/fcp.12858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Upcoming experimental and epidemiological data have identified the endogenous non-proteinogenic amino acid L-homoarginine (L-hArg) not only as a novel biomarker for cardiovascular disease but also as being directly involved in the pathogenesis of cardiac dysfunction. The association of low L-hArg levels with adverse cardiovascular events and mortality has proposed the idea of nutritional supplementation to rescue pathways inversely associated with cardiovascular health. Subsequent clinical and experimental studies contributed significantly to our knowledge of potential effects on the cardiorenal axis, acting either as a biomarker or a cardiovascular active agent. In this review article, we provide a comprehensive summary of the L-hArg metabolism, pathophysiological aspects, and current developments in the field of experimental and clinical evidence in favor of protective cardiovascular effects. Establishing a reliable biomarker to identify patients at high risk to die of cardiovascular disease represents one of the main goals for tackling this disease and providing individual therapeutic guidance.
Collapse
Affiliation(s)
- Vitali Koch
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.,Department of Cardiology, Angiology, and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | - Katrin Eichler
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - David M Leistner
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Christian Booz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Simon Bernatz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Tommaso D'Angelo
- Department of Biomedical Sciences and Morphological and Functional Imaging, University Hospital Messina, Messina, Italy
| | | | - Leona S Alizadeh
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Jan-Erik Scholtz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Ibrahim Yel
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Thomas J Vogl
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Winfried März
- Fifth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan E Hardt
- Department of Cardiology, Angiology, and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | - Simon S Martin
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
9
|
Cao G, Xuan X, Hu J, Zhang R, Jin H, Dong H. How vascular smooth muscle cell phenotype switching contributes to vascular disease. Cell Commun Signal 2022; 20:180. [PMID: 36411459 PMCID: PMC9677683 DOI: 10.1186/s12964-022-00993-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/22/2022] [Indexed: 11/22/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the most abundant cell in vessels. Earlier experiments have found that VSMCs possess high plasticity. Vascular injury stimulates VSMCs to switch into a dedifferentiated type, also known as synthetic VSMCs, with a high migration and proliferation capacity for repairing vascular injury. In recent years, largely owing to rapid technological advances in single-cell sequencing and cell-lineage tracing techniques, multiple VSMCs phenotypes have been uncovered in vascular aging, atherosclerosis (AS), aortic aneurysm (AA), etc. These VSMCs all down-regulate contractile proteins such as α-SMA and calponin1, and obtain specific markers and similar cellular functions of osteoblast, fibroblast, macrophage, and mesenchymal cells. This highly plastic phenotype transformation is regulated by a complex network consisting of circulating plasma substances, transcription factors, growth factors, inflammatory factors, non-coding RNAs, integrin family, and Notch pathway. This review focuses on phenotypic characteristics, molecular profile and the functional role of VSMCs phenotype landscape; the molecular mechanism regulating VSMCs phenotype switching; and the contribution of VSMCs phenotype switching to vascular aging, AS, and AA. Video Abstract.
Collapse
Affiliation(s)
- Genmao Cao
- grid.452845.a0000 0004 1799 2077Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Xuezhen Xuan
- grid.452845.a0000 0004 1799 2077Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Jie Hu
- grid.452845.a0000 0004 1799 2077Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Ruijing Zhang
- grid.452845.a0000 0004 1799 2077Department of Nephrology, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Haijiang Jin
- grid.452845.a0000 0004 1799 2077Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| | - Honglin Dong
- grid.452845.a0000 0004 1799 2077Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, No. 382, Wuyi Road, Taiyuan, China
| |
Collapse
|
10
|
Chakrabarti M, Bhattacharya A, Gebere MG, Johnson J, Ayub ZA, Chatzistamou I, Vyavahare NR, Azhar M. Increased TGFβ1 and SMAD3 Contribute to Age-Related Aortic Valve Calcification. Front Cardiovasc Med 2022; 9:770065. [PMID: 35928937 PMCID: PMC9343688 DOI: 10.3389/fcvm.2022.770065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 06/20/2022] [Indexed: 12/01/2022] Open
Abstract
Aims Calcific aortic valve disease (CAVD) is a progressive heart disease that is particularly prevalent in elderly patients. The current treatment of CAVD is surgical valve replacement, but this is not a permanent solution, and it is very challenging for elderly patients. Thus, a pharmacological intervention for CAVD may be beneficial. In this study, we intended to rescue aortic valve (AV) calcification through inhibition of TGFβ1 and SMAD3 signaling pathways. Methods and Results The klotho gene, which was discovered as an aging-suppressor gene, has been observed to play a crucial role in AV calcification. The klotho knockout (Kl–/–) mice have shorter life span (8–12 weeks) and develop severe AV calcification. Here, we showed that increased TGFβ1 and TGFβ-dependent SMAD3 signaling were associated with AV calcification in Kl–/– mice. Next, we generated Tgfb1- and Smad3-haploinsufficient Kl–/– mice to determine the contribution of TGFβ1 and SMAD3 to the AV calcification in Kl–/– mice. The histological and morphometric evaluation suggested a significant reduction of AV calcification in Kl–/–; Tgfb1± mice compared to Kl–/– mice. Smad3 heterozygous deletion was observed to be more potent in reducing AV calcification in Kl–/– mice compared to the Kl–/–; Tgfb1± mice. We observed significant inhibition of Tgfb1, Pai1, Bmp2, Alk2, Spp1, and Runx2 mRNA expression in Kl–/–; Tgfb1± and Kl–/–; Smad3± mice compared to Kl–/– mice. Western blot analysis confirmed that the inhibition of TGFβ canonical and non-canonical signaling pathways were associated with the rescue of AV calcification of both Kl–/–; Tgfb1± and Kl–/–; Smad3± mice. Conclusion Overall, inhibition of the TGFβ1-dependent SMAD3 signaling pathway significantly blocks the development of AV calcification in Kl–/– mice. This information is useful in understanding the signaling mechanisms involved in CAVD.
Collapse
Affiliation(s)
- Mrinmay Chakrabarti
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Aniket Bhattacharya
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, United States
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Mengistu G. Gebere
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - John Johnson
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Zeeshan A. Ayub
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | | | - Mohamad Azhar
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC, United States
- William Jennings Bryan Dorn VA Medical Center, Columbia, SC, United States
- *Correspondence: Mohamad Azhar,
| |
Collapse
|
11
|
Cao YC, Shan SK, Guo B, Li CC, Li FXZ, Zheng MH, Xu QS, Wang Y, Lei LM, Tang KX, Ou-Yang WL, Duan JY, Wu YY, Ullah MHE, Zhou ZA, Xu F, Lin X, Wu F, Liao XB, Yuan LQ. Histone Lysine Methylation Modification and Its Role in Vascular Calcification. Front Endocrinol (Lausanne) 2022; 13:863708. [PMID: 35784574 PMCID: PMC9243330 DOI: 10.3389/fendo.2022.863708] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/06/2022] [Indexed: 01/10/2023] Open
Abstract
Histone methylation is an epigenetic change mediated by histone methyltransferase, and has been connected to the beginning and progression of several diseases. The most common ailments that affect the elderly are cardiovascular and cerebrovascular disorders. They are the leading causes of death, and their incidence is linked to vascular calcification (VC). The key mechanism of VC is the transformation of vascular smooth muscle cells (VSMCs) into osteoblast-like phenotypes, which is a highly adjustable process involving a variety of complex pathophysiological processes, such as metabolic abnormalities, apoptosis, oxidative stress and signalling pathways. Many researchers have investigated the mechanism of VC and related targets for the prevention and treatment of cardiovascular and cerebrovascular diseases. Their findings revealed that histone lysine methylation modification may play a key role in the various stages of VC. As a result, a thorough examination of the role and mechanism of lysine methylation modification in physiological and pathological states is critical, not only for identifying specific molecular markers of VC and new therapeutic targets, but also for directing the development of new related drugs. Finally, we provide this review to discover the association between histone methylation modification and VC, as well as diverse approaches with which to investigate the pathophysiology of VC and prospective treatment possibilities.
Collapse
Affiliation(s)
- Ye-Chi Cao
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Su-Kang Shan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Guo
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chang-Chun Li
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fu-Xing-Zi Li
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Hui Zheng
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiu-Shuang Xu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Wang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li-Min Lei
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ke-Xin Tang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Lu Ou-Yang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Yue Duan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yun-Yun Wu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Muhammad Hasnain Ehsan Ullah
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Ang Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Wu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Bo Liao
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Qing Yuan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Ren SC, Mao N, Yi S, Ma X, Zou JQ, Tang X, Fan JM. Vascular Calcification in Chronic Kidney Disease: An Update and Perspective. Aging Dis 2022; 13:673-697. [PMID: 35656113 PMCID: PMC9116919 DOI: 10.14336/ad.2021.1024] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/24/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease is a devastating condition resulting from irreversible loss of nephron numbers and function and leading to end-stage renal disease and mineral disorders. Vascular calcification, an ectopic deposition of calcium-phosphate salts in blood vessel walls and heart valves, is an independent risk factor of cardiovascular morbidity and mortality in chronic kidney disease. Moreover, aging and related metabolic disorders are essential risk factors for chronic kidney disease and vascular calcification. Marked progress has been recently made in understanding and treating vascular calcification in chronic kidney disease. However, there is a paucity of systematic reviews summarizing this progress, and investigating unresolved issues is warranted. In this systematic review, we aimed to overview the underlying mechanisms of vascular calcification in chronic kidney diseases and discuss the impact of chronic kidney disease on the pathophysiology of vascular calcification. Additionally, we summarized potential clinical diagnostic biomarkers and therapeutic applications for vascular calcification with chronic kidney disease. This review may offer new insights into the pathogenesis, diagnosis, and therapeutic intervention of vascular calcification.
Collapse
Affiliation(s)
- Si-Chong Ren
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
- Center for Translational Medicine, Sichuan Academy of Traditional Chinese Medicine, Chengdu, China.
| | - Nan Mao
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Si Yi
- Chengdu Medical College, Chengdu, China.
- Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, China.
| | - Xin Ma
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Jia-Qiong Zou
- Chengdu Medical College, Chengdu, China.
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, Chengdu, China.
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jun-Ming Fan
- Chengdu Medical College, Chengdu, China.
- Clinical Research Center for Geriatrics of Sichuan Province, Chengdu, China.
| |
Collapse
|
13
|
Tsikas D, Redfors B. Pilot Study on Acute Effects of Pharmacological Intraperitoneal L-Homoarginine on Homeostasis of Lysine and Other Amino Acids in a Rat Model of Isoprenaline-Induced Takotsubo Cardiomyopathy. Int J Mol Sci 2022; 23:ijms23094734. [PMID: 35563125 PMCID: PMC9103764 DOI: 10.3390/ijms23094734] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/04/2023] Open
Abstract
L-Arginine:glycine amidinotransferase (AGAT) catalyzes the formation of L-homoarginine (hArg) and L-ornithine (Orn) from L-arginine (Arg) and L-lysine (Lys): Arg + Lys ↔ hArg + Orn; equilibrium constant KhArg. AGAT also catalyzes the formation of guanidinoacetate (GAA) and Orn from Arg and glycine (Gly): Arg + Gly ↔ GAA + Orn; equilibrium constant KGAA. In humans, pharmacological hArg is metabolized to Lys. Low circulating and low excretory concentrations of hArg are associated with worse outcomes and mortality in the renal and cardiovascular systems. The metabolism and pharmacology of hArg have been little investigated. In the present study, we investigated the effects of pharmacological hArg (i.p., 0, 20, 220, 440 mg/kg at time point 0 min) on amino acids homeostasis in a rat model of isoprenaline-induced takotsubo cardiomyopathy (i.p., 50 mg/kg at time point 15 min). We measured by gas chromatography-mass spectrometry free and proteinic amino acids, as well as the polyamines putrescine and spermidine in the heart, lung, kidney, and liver of ten rats sacrificed at various time points (range, 0 to 126 min). hArg administration resulted in multiple changes in the tissue contents of several free and proteinic amino acids, as well as in the putrescine-spermidine molar ratio, an indicator of polyamines catabolism. Our results suggest that Lys and Arg are major metabolites of pharmacological hArg. Kidneys and heart seem to play a major metabolic role for hArg. Circulating Lys does not change over time, yet there is a considerable interchange of free Lys between organs, notably kidney and heart, during the presence of isoprenaline in the rats (time range, 15 to 90 min). Antidromic changes were observed for KhArg and KGAA, notably in the heart in this time window. Our study shows for the first time that free hArg and sarcosine (N-methylglycine) are positively associated with each other. The acute effects of high-dosed hArg administration and isoprenaline on various amino acids and on AGAT-catalyzed reaction in the heart, lung, kidney, and liver are detailed and discussed.
Collapse
Affiliation(s)
- Dimitrios Tsikas
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, 30623 Hannover, Germany
- Correspondence:
| | - Björn Redfors
- Department of Cardiology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden;
| |
Collapse
|
14
|
Activating BK channels ameliorates vascular smooth muscle calcification through Akt signaling. Acta Pharmacol Sin 2022; 43:624-633. [PMID: 34163023 PMCID: PMC8888620 DOI: 10.1038/s41401-021-00704-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Vascular calcification (VC) is characterized by pathological depositions of calcium and phosphate in the arteries and veins via an active cell-regulated process, in which vascular smooth muscle cells (VSMCs) transform into osteoblast/chondrocyte-like cells as in bone formation. VC is associated with significant morbidity and mortality in chronic kidney disease (CKD) and cardiovascular disease, but the underlying mechanisms remain unclear. In this study we investigated the role of large-conductance calcium-activated potassium (BK) channels in 3 experimental VC models. VC was induced in vascular smooth muscle cells (VSMCs) by β-glycerophosphate (β-GP), or in rats by subtotal nephrectomy, or in mice by high-dosage vitamin D3. We showed that the expression of BK channels in the artery of CKD rats with VC and in β-GP-treated VSMCs was significantly decreased, which was functionally confirmed by patch-clamp recording. In β-GP-treated VSMCs, BK channel opener NS1619 (20 μM) significantly alleviated VC by decreasing calcium content and alkaline phosphatase activity. Furthermore, NS1619 decreased mRNA expression of ostoegenic genes OCN and OPN, as well as Runx2 (a key transcription factor involved in preosteoblast to osteoblast differentiation), and increased the expression of α-SMA protein, whereas BK channel inhibitor paxilline (10 μM) caused the opposite effects. In primary cultured VSMCs from BK-/- mice, BK deficiency aggravated calcification as did BK channel inhibitor in normal VSMCs. Moreover, calcification was more severe in thoracic aorta rings of BK-/- mice than in those of wild-type littermates. Administration of BK channel activator BMS191011 (10 mg· kg-1 ·d-1) in high-dosage vitamin D3-treated mice significantly ameliorated calcification. Finally, co-treatment with Akt inhibitor MK2206 (1 μM) or FoxO1 inhibitor AS1842856 (3 μM) in calcified VSMCs abrogated the effects of BK channel opener NS1619. Taken together, activation of BK channels ameliorates VC via Akt/FoxO1 signaling pathways. Strategies to activate BK channels and/or enhance BK channel expression may offer therapeutic avenues to control VC.
Collapse
|
15
|
Hu MC, Moe OW. Phosphate and Cellular Senescence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:55-72. [PMID: 35288873 PMCID: PMC10513121 DOI: 10.1007/978-3-030-91623-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cellular senescence is one type of permeant arrest of cell growth and one of increasingly recognized contributor to aging and age-associated disease. High phosphate and low Klotho individually and synergistically lead to age-related degeneration in multiple organs. Substantial evidence supports the causality of high phosphate in cellular senescence, and potential contribution to human aging, cancer, cardiovascular, kidney, neurodegenerative, and musculoskeletal diseases. Phosphate can induce cellular senescence both by direct phosphotoxicity, and indirectly through downregulation of Klotho and upregulation of plasminogen activator inhibitor-1. Restriction of dietary phosphate intake and blockage of intestinal absorption of phosphate help suppress cellular senescence. Supplementation of Klotho protein, cellular senescence inhibitor, and removal of senescent cells with senolytic agents are potential novel strategies to attenuate phosphate-induced cellular senescence, retard aging, and ameliorate age-associated, and phosphate-induced disorders.
Collapse
Affiliation(s)
- Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
16
|
Protective effects of spironolactone on vascular calcification in chronic kidney disease. Biochem Biophys Res Commun 2021; 582:28-34. [PMID: 34678593 DOI: 10.1016/j.bbrc.2021.10.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/09/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Vascular calcification is common in chronic kidney disease (CKD) and associated with increased cardiovascular mortality. Aldosterone has been implicated as an augmenting factor in the progression of vascular calcification. The present study further explored putative beneficial effects of aldosterone inhibition by the mineralocorticoid receptor antagonist spironolactone on vascular calcification in CKD. METHODS Serum calcification propensity was determined in serum samples from the MiREnDa trial, a prospective, randomized controlled clinical trial to investigate efficacy and safety of spironolactone in maintenance hemodialysis patients. Experiments were conducted in mice with subtotal nephrectomy and cholecalciferol treatment, and in calcifying primary human aortic smooth muscle cells (HAoSMCs). RESULTS Serum calcification propensity was improved by spironolactone treatment in patients on hemodialysis from the MiREnDa trial. In mouse models and HAoSMCs, spironolactone treatment ameliorated vascular calcification and expression of osteogenic markers. CONCLUSIONS These observations support a putative benefit of spironolactone treatment in CKD-associated vascular calcification. Further research is required to investigate possible improvements in cardiovascular outcomes by spironolactone and whether the benefits outweigh the risks in patients with CKD.
Collapse
|
17
|
Henze LA, Estepa M, Pieske B, Lang F, Eckardt KU, Alesutan I, Voelkl J. Zinc Ameliorates the Osteogenic Effects of High Glucose in Vascular Smooth Muscle Cells. Cells 2021; 10:cells10113083. [PMID: 34831306 PMCID: PMC8623153 DOI: 10.3390/cells10113083] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/23/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
In diabetic patients, medial vascular calcification is common and associated with increased cardiovascular mortality. Excessive glucose concentrations can activate the nuclear factor kappa-light-chain-enhancer of activated B-cells (NF-kB) and trigger pro-calcific effects in vascular smooth muscle cells (VSMCs), which may actively augment vascular calcification. Zinc is able to mitigate phosphate-induced VSMC calcification. Reduced serum zinc levels have been reported in diabetes mellitus. Therefore, in this study the effects of zinc supplementation were investigated in primary human aortic VSMCs exposed to excessive glucose concentrations. Zinc treatment was found to abrogate the stimulating effects of high glucose on VSMC calcification. Furthermore, zinc was found to blunt the increased expression of osteogenic and chondrogenic markers in high glucose-treated VSMCs. High glucose exposure was shown to activate NF-kB in VSMCs, an effect that was blunted by additional zinc treatment. Zinc was further found to increase the expression of TNFα-induced protein 3 (TNFAIP3) in high glucose-treated VSMCs. The silencing of TNFAIP3 was shown to abolish the protective effects of zinc on high glucose-induced NF-kB-dependent transcriptional activation, osteogenic marker expression, and the calcification of VSMCs. Silencing of the zinc-sensing receptor G protein-coupled receptor 39 (GPR39) was shown to abolish zinc-induced TNFAIP3 expression and the effects of zinc on high glucose-induced osteogenic marker expression. These observations indicate that zinc may be a protective factor during vascular calcification in hyperglycemic conditions.
Collapse
Affiliation(s)
- Laura A. Henze
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany; (L.A.H.); (M.E.); (B.P.)
| | - Misael Estepa
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany; (L.A.H.); (M.E.); (B.P.)
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum, 13353 Berlin, Germany; (L.A.H.); (M.E.); (B.P.)
| | - Florian Lang
- Department of Vegetative and Clinical Physiology, Eberhard Karls University Tübingen, 72076 Tübingen, Germany;
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.-U.E.); (J.V.)
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, 4040 Linz, Austria
- Correspondence: ; Tel.: +43-732-2468-8990
| | - Jakob Voelkl
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (K.-U.E.); (J.V.)
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, 4040 Linz, Austria
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347 Berlin, Germany
| |
Collapse
|
18
|
Acid sphingomyelinase promotes SGK1-dependent vascular calcification. Clin Sci (Lond) 2021; 135:515-534. [PMID: 33479769 PMCID: PMC7859357 DOI: 10.1042/cs20201122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/07/2021] [Accepted: 01/21/2021] [Indexed: 12/20/2022]
Abstract
In chronic kidney disease (CKD), hyperphosphatemia is a key factor promoting medial vascular calcification, a common complication associated with cardiovascular events and high mortality. Vascular calcification involves osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs), but the complex signaling events inducing pro-calcific pathways are incompletely understood. The present study investigated the role of acid sphingomyelinase (ASM)/ceramide as regulator of VSMC calcification. In vitro, both, bacterial sphingomyelinase and phosphate increased ceramide levels in VSMCs. Bacterial sphingomyelinase as well as ceramide supplementation stimulated osteo-/chondrogenic transdifferentiation during control and high phosphate conditions and augmented phosphate-induced calcification of VSMCs. Silencing of serum- and glucocorticoid-inducible kinase 1 (SGK1) blunted the pro-calcific effects of bacterial sphingomyelinase or ceramide. Asm deficiency blunted vascular calcification in a cholecalciferol-overload mouse model and ex vivo isolated-perfused arteries. In addition, Asm deficiency suppressed phosphate-induced osteo-/chondrogenic signaling and calcification of cultured VSMCs. Treatment with the functional ASM inhibitors amitriptyline or fendiline strongly blunted pro-calcific signaling pathways in vitro and in vivo. In conclusion, ASM/ceramide is a critical upstream regulator of vascular calcification, at least partly, through SGK1-dependent signaling. Thus, ASM inhibition by repurposing functional ASM inhibitors to reduce the progression of vascular calcification during CKD warrants further study.
Collapse
|
19
|
Mokhaneli MC, Botha-Le Roux S, Fourie CMT, Böger R, Schwedhelm E, Mels CMC. L-homoarginine is associated with decreased cardiovascular- and all-cause mortality. Eur J Clin Invest 2021; 51:e13472. [PMID: 33320332 DOI: 10.1111/eci.13472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Increasing evidence suggests that L-homoarginine, an endogenous analogue of the amino acid L-arginine, may have beneficial effects on vascular homeostasis. We examined whether L-homoarginine is associated with 10-year risk of all-cause and cardiovascular mortality in a black South African population. METHODS We included 669 black South African participants (mean age 59.5 years), 143 of whom died during the 10-year follow-up period. Mortality data were acquired via verbal autopsy. Plasma L-homoarginine (and other related markers) were analysed with liquid chromatography-tandem mass spectrometry. RESULTS Survivors had higher L-homoarginine levels compared with nonsurvivors (1.25 µM vs. 0.89 µM; P < .001). Multivariable Cox regression analyses revealed that higher plasma L-homoarginine predicted a reduction in 10-year cardiovascular (hazard ratio [HR] per SD increment, 0.61; 95% CI 0.50 to 0.75) and all-cause (hazard ratio [HR] per SD increment, 0.59; 95% CI 0.41 to 0.84) mortality risk. CONCLUSION Higher L-homoarginine levels are associated with reduced risk of 10-year cardiovascular and all-cause mortality. Regulation of L-homoarginine levels as a therapeutic target in the management of cardiovascular disease should be investigated.
Collapse
Affiliation(s)
- Maserame Cleopatra Mokhaneli
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Shani Botha-Le Roux
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Carla Maria Theresia Fourie
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Rainer Böger
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Deutsches Zentrum fuer Herz-Kreislauf-Forschung E.V. (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Edzard Schwedhelm
- Department of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Deutsches Zentrum fuer Herz-Kreislauf-Forschung E.V. (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Catharina Martha Cornelia Mels
- Faculty of Health Sciences, Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| |
Collapse
|
20
|
Alesutan I, Luong TTD, Schelski N, Masyout J, Hille S, Schneider MP, Graham D, Zickler D, Verheyen N, Estepa M, Pasch A, Maerz W, Tomaschitz A, Pilz S, Frey N, Lang F, Delles C, Müller OJ, Pieske B, Eckardt KU, Scherberich J, Voelkl J. Circulating uromodulin inhibits vascular calcification by interfering with pro-inflammatory cytokine signalling. Cardiovasc Res 2021; 117:930-941. [PMID: 32243494 DOI: 10.1093/cvr/cvaa081] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/15/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022] Open
Abstract
AIMS Uromodulin is produced exclusively in the kidney and secreted into both urine and blood. Serum levels of uromodulin are correlated with kidney function and reduced in chronic kidney disease (CKD) patients, but physiological functions of serum uromodulin are still elusive. This study investigated the role of uromodulin in medial vascular calcification, a key factor associated with cardiovascular events and mortality in CKD patients. METHODS AND RESULTS Experiments were performed in primary human (HAoSMCs) and mouse (MOVAS) aortic smooth muscle cells, cholecalciferol overload and subtotal nephrectomy mouse models and serum from CKD patients. In three independent cohorts of CKD patients, serum uromodulin concentrations were inversely correlated with serum calcification propensity. Uromodulin supplementation reduced phosphate-induced osteo-/chondrogenic transdifferentiation and calcification of HAoSMCs. In human serum, pro-inflammatory cytokines tumour necrosis factor α (TNFα) and interleukin-1β (IL-1β) co-immunoprecipitated with uromodulin. Uromodulin inhibited TNFα and IL-1β-induced osteo-/chondrogenic signalling and activation of the transcription factor nuclear factor kappa-light-chain-enhancer of activated β cells (NF-kB) as well as phosphate-induced NF-kB-dependent transcriptional activity in HAoSMCs. In vivo, adeno-associated virus (AAV)-mediated overexpression of uromodulin ameliorated vascular calcification in mice with cholecalciferol overload. Conversely, cholecalciferol overload-induced vascular calcification was aggravated in uromodulin-deficient mice. In contrast, uromodulin overexpression failed to reduce vascular calcification during renal failure in mice. Carbamylated uromodulin was detected in serum of CKD patients and uromodulin carbamylation inhibited its anti-calcific properties in vitro. CONCLUSIONS Uromodulin counteracts vascular osteo-/chondrogenic transdifferentiation and calcification, at least in part, through interference with cytokine-dependent pro-calcific signalling. In CKD, reduction and carbamylation of uromodulin may contribute to vascular pathology.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Aorta/immunology
- Aorta/metabolism
- Cell Transdifferentiation/drug effects
- Cells, Cultured
- Chondrogenesis
- Cytokines/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Humans
- Inflammation Mediators/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Middle Aged
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- Osteogenesis
- Phenotype
- Protein Carbamylation
- Renal Insufficiency, Chronic/blood
- Renal Insufficiency, Chronic/immunology
- Signal Transduction
- Uromodulin/blood
- Uromodulin/genetics
- Uromodulin/pharmacology
- Vascular Calcification/blood
- Vascular Calcification/immunology
- Vascular Calcification/prevention & control
- Young Adult
- Mice
Collapse
Affiliation(s)
- Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch 2, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
| | - Trang T D Luong
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nadeshda Schelski
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Jaber Masyout
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Susanne Hille
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105 Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Martinistr. 52, 20246 Hamburg, Germany
| | - Markus P Schneider
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
- German Chronic Kidney Disease (GCKD) Study
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Daniel Zickler
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nicolas Verheyen
- Department of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Misael Estepa
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andreas Pasch
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
- Calciscon AG, Aarbergstrasse 5, 2560 Nidau-Biel, Switzerland
- Nierenpraxis Bern, Bubenbergplatz 5, 3011 Bern, Switzerland
- Department of Nephrology, Lindenhofspital, Bremgartenstrasse 117, 3001 Bern, Switzerland
| | - Winfried Maerz
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
- Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Ludolf Krehl Street 7-11, 68167 Mannheim, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, P5,7, 68161 Mannheim, Germany
| | | | - Stefan Pilz
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Norbert Frey
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105 Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Martinistr. 52, 20246 Hamburg, Germany
| | - Florian Lang
- Department of Physiology, Eberhard-Karls University, Wilhelmstr. 56, 72076 Tübingen, Germany
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Str. 3, 24105 Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Martinistr. 52, 20246 Hamburg, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch 2, 10178 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
- Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
- German Chronic Kidney Disease (GCKD) Study
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Juergen Scherberich
- Department of Nephrology and Clinical Immunology, Klinikum München-Harlaching, Teaching Hospital of the Ludwig-Maximilians-Universität, Sanatoriumsplatz 2, 81545 München, Germany
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040 Linz, Austria
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Hessische Strasse 3-4, 10115 Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
21
|
Lee HY, Lim S, Park S. Role of Inflammation in Arterial Calcification. Korean Circ J 2021; 51:114-125. [PMID: 33525066 PMCID: PMC7853899 DOI: 10.4070/kcj.2020.0517] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 12/24/2020] [Indexed: 01/11/2023] Open
Abstract
Arterial calcification, characterized by calcium phosphate deposition in the arteries, can be divided into intimal calcification and medial calcification. The former is the predominant form of calcification in coronary artery plaques; the latter mostly affects peripheral arteries and aortas. Both forms of arterial calcification have strong correlations with adverse cardiovascular events. Intimal microcalcification is associated with increased risk of plaque disruption while the degree of burden of coronary calcification, measured by coronary calcium score, is a marker of overall plaque burden. Continuous research on vascular calcification has been performed during the past few decades, and several cellular and molecular mechanisms and therapeutic targets were identified. However, despite clinical trials to evaluate the efficacy of drug therapies to treat vascular calcification, none have been shown to have efficacy until the present. Therefore, more extensive research is necessary to develop appropriate therapeutic strategies based on a thorough understanding of vascular calcification. In this review, we mainly focus on intimal calcification, namely the pathobiology of arterial calcification, and its clinical implications.
Collapse
Affiliation(s)
- Hae Young Lee
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, Seoul, Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Korea
| | - Sungha Park
- Division of Cardiology, Severance Cardiovascular Hospital and Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
22
|
Oe Y, Mitsui S, Sato E, Shibata N, Kisu K, Sekimoto A, Miyazaki M, Sato H, Ito S, Takahashi N. Lack of Endothelial Nitric Oxide Synthase Accelerates Ectopic Calcification in Uremic Mice Fed an Adenine and High Phosphorus Diet. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:283-293. [PMID: 33159888 DOI: 10.1016/j.ajpath.2020.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/08/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022]
Abstract
Ectopic calcification is a risk of cardiovascular disease in chronic kidney disease (CKD) patients, and impaired endothelial nitric oxide synthase (eNOS) is involved in the CKD complications. However, whether eNOS dysfunction is a cause of ectopic calcification in CKD remains to be elucidated. To address this issue, we investigated the role of eNOS in ectopic calcification in mice with renal injury caused by an adenine and high-phosphorus (Ade + HP) diet. DBA/2J mice, a calcification-sensitive strain, were fed Ade + HP for 3 weeks. Expression levels of eNOS-related genes were reduced significantly in their calcified aorta. C57BL/6J is a calcification-resistant strain, and wild-type mice showed mild calcified lesions in the aorta and kidney when given an Ade + HP diet for 4 weeks. In contrast, a lack of eNOS led to the development of severe aortic calcification accompanied by an increase in runt-related transcription factor 2, an osteochondrogenic marker. Increased renal calcium deposition and the tubular injury score were remarkable in mice lacking eNOS-fed Ade + HP. Exacerbation of ectopic calcification by a lack of eNOS is associated with increased oxidative stress markers such as nicotinamide adenine dinucleotide phosphate oxidases. In conclusion, eNOS is critically important in preventing ectopic calcification. Therefore, the maintenance of eNOS is useful to reduce cardiovascular disease events and to improve prognosis in CKD patients.
Collapse
Affiliation(s)
- Yuji Oe
- Department of Community Medical Support, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Shohei Mitsui
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan
| | - Emiko Sato
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan
| | - Naoko Shibata
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kiyomi Kisu
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akiyo Sekimoto
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan
| | - Mariko Miyazaki
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Sato
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan
| | - Sadayoshi Ito
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuyuki Takahashi
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan; Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Faculty of Pharmaceutical Sciences, Sendai, Japan.
| |
Collapse
|
23
|
Bao S, Guo Y, Diao Z, Guo W, Liu W. Genome-wide identification of lncRNAs and mRNAs differentially expressed in human vascular smooth muscle cells stimulated by high phosphorus. Ren Fail 2020; 42:437-446. [PMID: 32401115 PMCID: PMC7269060 DOI: 10.1080/0886022x.2020.1758722] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Cardiovascular events are the primary cause of death for chronic kidney disease patients, which occurred via vascular calcification evolving pathogenically. Although a high level of phosphorus contributes to the induction of osteogenic differentiation of vascular smooth muscle cells (VSMCs), the role of lncRNA in this process awaits further study.Methods: In this study, we systematically investigated the variation of gene expression in human VSMCs induced by high phosphorus. LncRNAs and mRNAs expression were revealed by microarray analyses of the control group and high-phosphorus (HP) group. LncRNA-mRNA co-expression network was established based on the specific lncRNA-mRNA relationships. Hierarchical clustering was used to identify a common set of regulated genes. In addition, Gene Ontology enrichment, Kyoto Gene-Encyclopedia and genomic analyses were conducted for the mRNAs differentially expressed under high phosphorus.Result: RT-qPCR results confirmed that the expression of RUNX2, BMP2 and osteocalcin in HP group exhibited significant increases than in control group (p < .05). VSMC in HP group also showed higher intracellular calcium content. Volcano plots results show that 379 mRNAs and 728 lncRNAs different expressed in HP group. LncRNA-mRNA co-expression networks analysis revealed that 8 lncRNAs were the most highly connected lncRNAs. Quantitative analysis indicated that two lncRNAs were confirmed to increase significantly in the HP group. The mRNA expression of NT5E and ICAM1 were higher in group HP, while MAP3K7CL was lower than CON group (p < .05).Conclusion: This study provided a working list of lncRNAs that may be relevant to osteogenic differentiation, which presents a new insights into the mechanism of vascular calcification induced by high phosphorus in VSMCs.
Collapse
MESH Headings
- 5'-Nucleotidase/genetics
- Cell Line
- GPI-Linked Proteins/genetics
- Gene Expression
- Gene Expression Profiling
- Gene Ontology
- Humans
- Intercellular Adhesion Molecule-1/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Osteogenesis
- Phosphorus/metabolism
- Protein Kinases/genetics
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
Collapse
Affiliation(s)
- Shumin Bao
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing, China
| | - Yan Guo
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing, China
| | - Zongli Diao
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing, China
| | - Weikang Guo
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing, China
| | - Wenhu Liu
- Department of Nephrology, Beijing Friendship Hospital, Faculty of Kidney Diseases, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Regulation of Vascular Calcification by Reactive Oxygen Species. Antioxidants (Basel) 2020; 9:antiox9100963. [PMID: 33049989 PMCID: PMC7599480 DOI: 10.3390/antiox9100963] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/18/2022] Open
Abstract
Vascular calcification is the deposition of hydroxyapatite crystals in the medial or intimal layers of arteries that is usually associated with other pathological conditions including but not limited to chronic kidney disease, atherosclerosis and diabetes. Calcification is an active, cell-regulated process involving the phenotype transition of vascular smooth muscle cells (VSMCs) from contractile to osteoblast/chondrocyte-like cells. Diverse triggers and signal transduction pathways have been identified behind vascular calcification. In this review, we focus on the role of reactive oxygen species (ROS) in the osteochondrogenic phenotype switch of VSMCs and subsequent calcification. Vascular calcification is associated with elevated ROS production. Excessive ROS contribute to the activation of certain osteochondrogenic signal transduction pathways, thereby accelerating osteochondrogenic transdifferentiation of VSMCs. Inhibition of ROS production and ROS scavengers and activation of endogenous protective mechanisms are promising therapeutic approaches in the prevention of osteochondrogenic transdifferentiation of VSMCs and subsequent vascular calcification. The present review discusses the formation and actions of excess ROS in different experimental models of calcification, and the potential of ROS-lowering strategies in the prevention of this deleterious condition.
Collapse
|
25
|
Shi J, Yang Y, Cheng A, Xu G, He F. Metabolism of vascular smooth muscle cells in vascular diseases. Am J Physiol Heart Circ Physiol 2020; 319:H613-H631. [PMID: 32762559 DOI: 10.1152/ajpheart.00220.2020] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are the fundamental component of the medial layer of arteries and are essential for arterial physiology and pathology. It is becoming increasingly clear that VSMCs can alter their metabolism to fulfill the bioenergetic and biosynthetic requirements. During vascular injury, VSMCs switch from a quiescent "contractile" phenotype to a highly migratory and proliferative "synthetic" phenotype. Recent studies have found that the phenotype switching of VSMCs is driven by a metabolic switch. Metabolic pathways, including aerobic glycolysis, fatty acid oxidation, and amino acid metabolism, have distinct, indispensable roles in normal and dysfunctional vasculature. VSMCs metabolism is also related to the metabolism of endothelial cells. In the present review, we present a brief overview of VSMCs metabolism and how it regulates the progression of several vascular diseases, including atherosclerosis, systemic hypertension, diabetes, pulmonary hypertension, vascular calcification, and aneurysms, and the effect of the risk factors for vascular disease (aging, cigarette smoking, and excessive alcohol drinking) on VSMC metabolism to clarify the role of VSMCs metabolism in the key pathological process.
Collapse
Affiliation(s)
- Jia Shi
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anying Cheng
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Xu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan He
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Bao JF, Hu PP, She QY, Li A. A Land of Controversy: Fibroblast Growth Factor-23 and Uremic Cardiac Hypertrophy. J Am Soc Nephrol 2020; 31:1423-1434. [PMID: 32527977 PMCID: PMC7351013 DOI: 10.1681/asn.2020010081] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiac hypertrophy is a common feature in patients with CKD. Recent studies revealed that two phosphate regulators, fibroblast growth factor-23 and α-Klotho, are highly involved in the pathophysiologic process of CKD-induced cardiac hypertrophy. With decreasing renal function, elevated fibroblast growth factor-23 and decreased α-Klotho may contribute to cardiac hypertrophy by targeting the heart directly or by inducing systemic changes, such as vascular injury, hemodynamic disorders, and inflammation. However, several studies have demonstrated that disturbances in the fibroblast growth factor-23/α-Klotho axis do not lead to cardiac hypertrophy. In this review, we describe the cardiac effects of the fibroblast growth factor-23/α-Klotho axis and summarize recent progress in this field. In addition, we present not only the main controversies in this field but also provide possible directions to resolve these disputes.
Collapse
Affiliation(s)
- Jing-Fu Bao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pan-Pan Hu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qin-Ying She
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Aiqing Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Varennes O, Mentaverri R, Duflot T, Kauffenstein G, Objois T, Lenglet G, Avondo C, Morisseau C, Brazier M, Kamel S, Six I, Bellien J. The Metabolism of Epoxyeicosatrienoic Acids by Soluble Epoxide Hydrolase Is Protective against the Development of Vascular Calcification. Int J Mol Sci 2020; 21:ijms21124313. [PMID: 32560362 PMCID: PMC7352784 DOI: 10.3390/ijms21124313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
This study addressed the hypothesis that soluble epoxide hydrolase (sEH), which metabolizes endothelium-derived epoxyeicosatrienoic acids, plays a role in vascular calcification. The sEH inhibitor trans-4-(4-(3-adamantan-1-yl-ureido)-cyclohexyloxy)-benzoic acid (t-AUCB) potentiated the increase in calcium deposition of rat aortic rings cultured in high-phosphate conditions. This was associated with increased tissue-nonspecific alkaline phosphatase activity and mRNA expression level of the osteochondrogenic marker Runx2. The procalcifying effect of t-AUCB was prevented by mechanical aortic deendothelialization or inhibition of the production and action of epoxyeicosatrienoic acids using the cytochrome P450 inhibitor fluconazole and the antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE), respectively. Similarly, exogenous epoxyeicosatrienoic acids potentiated the calcification of rat aortic rings through a protein kinase A (PKA)-dependent mechanism and of human aortic vascular smooth muscle cells when sEH was inhibited by t-AUCB. Finally, a global gene expression profiling analysis revealed that the mRNA expression level of sEH was decreased in human carotid calcified plaques compared to adjacent lesion-free sites and was inversely correlated with Runx2 expression. These results show that sEH hydrolase plays a protective role against vascular calcification by reducing the bioavailability of epoxyeicosatrienoic acids.
Collapse
Affiliation(s)
- Olivier Varennes
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
| | - Romuald Mentaverri
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
- Department of Biochemistry, Amiens-Picardie University Hospital, 80054 Amiens, France
| | - Thomas Duflot
- Department of Pharmacology, Rouen University Hospital, CEDEX 1, 76031 Rouen, France;
- INSERM U1096, Normandy University, UNIROUEN, F-76000 Rouen, France
| | | | - Thibaut Objois
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
| | - Gaëlle Lenglet
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
| | - Carine Avondo
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
| | - Christophe Morisseau
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA 95616, USA;
| | - Michel Brazier
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
| | - Saïd Kamel
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
- Department of Biochemistry, Amiens-Picardie University Hospital, 80054 Amiens, France
| | - Isabelle Six
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
- Correspondence: (I.S.); (J.B.); Tel.: +33-2-32-88-90-30 (J.B.); Fax: +33-2-32-88-91-16 (J.B)
| | - Jeremy Bellien
- Department of Pharmacology, Rouen University Hospital, CEDEX 1, 76031 Rouen, France;
- INSERM U1096, Normandy University, UNIROUEN, F-76000 Rouen, France
- Correspondence: (I.S.); (J.B.); Tel.: +33-2-32-88-90-30 (J.B.); Fax: +33-2-32-88-91-16 (J.B)
| |
Collapse
|
28
|
Lee SJ, Lee IK, Jeon JH. Vascular Calcification-New Insights Into Its Mechanism. Int J Mol Sci 2020; 21:ijms21082685. [PMID: 32294899 PMCID: PMC7216228 DOI: 10.3390/ijms21082685] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification (VC), which is categorized by intimal and medial calcification, depending on the site(s) involved within the vessel, is closely related to cardiovascular disease. Specifically, medial calcification is prevalent in certain medical situations, including chronic kidney disease and diabetes. The past few decades have seen extensive research into VC, revealing that the mechanism of VC is not merely a consequence of a high-phosphorous and -calcium milieu, but also occurs via delicate and well-organized biologic processes, including an imbalance between osteochondrogenic signaling and anticalcific events. In addition to traditionally established osteogenic signaling, dysfunctional calcium homeostasis is prerequisite in the development of VC. Moreover, loss of defensive mechanisms, by microorganelle dysfunction, including hyper-fragmented mitochondria, mitochondrial oxidative stress, defective autophagy or mitophagy, and endoplasmic reticulum (ER) stress, may all contribute to VC. To facilitate the understanding of vascular calcification, across any number of bioscientific disciplines, we provide this review of a detailed updated molecular mechanism of VC. This encompasses a vascular smooth muscle phenotypic of osteogenic differentiation, and multiple signaling pathways of VC induction, including the roles of inflammation and cellular microorganelle genesis.
Collapse
Affiliation(s)
- Sun Joo Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea;
| | - In-Kyu Lee
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea;
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Jae-Han Jeon
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Korea;
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: ; Tel.: +82-(53)-200-3182; Fax: +82-(53)-200-3155
| |
Collapse
|
29
|
An overview of the mechanisms in vascular calcification during chronic kidney disease. Curr Opin Nephrol Hypertens 2020; 28:289-296. [PMID: 30985336 DOI: 10.1097/mnh.0000000000000507] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Chronic kidney disease (CKD) facilitates a unique environment to strongly accelerate vascular calcification - the pathological deposition of calcium-phosphate in the vasculature. These calcifications are associated with the excessive cardiovascular mortality of CKD patients. RECENT FINDINGS Vascular calcification is a multifaceted active process, mediated, at least partly, by vascular smooth muscle cells. These cells are able to transdifferentiate into cells with osteo/chondrogenic properties, which exert multiple effects to facilitate vascular tissue mineralization. As the understanding of the underlying pathophysiology increases, first therapeutic concepts begin to emerge. SUMMARY This brief review provides an overview on the so far known mechanisms involved in the initiation and progression of vascular calcification in CKD.
Collapse
|
30
|
Sasani A, Hornig S, Grzybowski R, Cordts K, Hanff E, Tsikas D, Böger R, Gerloff C, Isbrandt D, Neu A, Schwedhelm E, Choe CU. Muscle phenotype of AGAT- and GAMT-deficient mice after simvastatin exposure. Amino Acids 2019; 52:73-85. [DOI: 10.1007/s00726-019-02812-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 12/05/2019] [Indexed: 01/03/2023]
|
31
|
Synthesis of homoagmatine and GC–MS analysis of tissue homoagmatine and agmatine: evidence that homoagmatine but not agmatine is a metabolite of pharmacological L-homoarginine in the anesthetized rat. Amino Acids 2019; 52:235-245. [DOI: 10.1007/s00726-019-02808-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023]
|
32
|
Hanff E, Said MY, Kayacelebi AA, Post A, Minovic I, van den Berg E, de Borst MH, van Goor H, Bakker SJL, Tsikas D. High plasma guanidinoacetate-to-homoarginine ratio is associated with high all-cause and cardiovascular mortality rate in adult renal transplant recipients. Amino Acids 2019; 51:1485-1499. [PMID: 31535220 DOI: 10.1007/s00726-019-02783-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 08/30/2019] [Indexed: 12/11/2022]
Abstract
L-Arginine:glycine amidinotransferase (AGAT) is the main producer of the creatine precursor, guanidinoacetate (GAA), and L-homoarginine (hArg). We and others previously reported lower levels of circulating and urinary hArg in renal transplant recipients (RTR) compared to healthy subjects. In adults, hArg emerged as a novel risk factor for renal and cardiovascular adverse outcome. Urinary GAA was found to be lower in children and adolescents with kidney transplants compared to healthy controls. Whether GAA is also a risk factor in the renal and cardiovascular systems of adults, is not yet known. In the present study, we aimed to investigate the significance of circulating GAA and the GAA-to-hArg molar ratio (GAA/hArg) in adult RTR. We hypothesized that GAA/hArg represents a measure of the balanced state of the AGAT activity in the kidneys, and would prospectively allow assessing a potential association between GAA/hArg and long-term outcome in RTR. The median follow-up period was 5.4 years. Confounders and potential mediators of GAA/hArg associations were evaluated with multivariate linear regression analyses, and the association with all-cause and cardiovascular mortality or death-censored graft loss was studied with Cox regression analyses. The study cohort consisted of 686 stable RTR and 140 healthy kidney donors. Median plasma GAA concentration was significantly lower in the RTR compared to the kidney donors before kidney donation: 2.19 [1.77-2.70] µM vs. 2.78 [2.89-3.35] µM (P < 0.001). In cross-sectional multivariable analyses in RTR, HDL cholesterol showed the strongest association with GAA/hArg. In prospective analyses in RTR, GAA/hArg was associated with a higher risk for all-cause mortality (hazard ratio (HR): 1.35 [95% CI 1.19-1.53]) and cardiovascular mortality (HR: 1.46 [95% CI 1.24-1.73]), independent of potential confounders. GAA but not GAA/hArg was associated with death-censored graft loss in crude survival and Cox regression analyses. The association of GAA and death-censored graft loss was lost after adjustment for eGFR. Our study suggests that in the kidneys of RTR, the AGAT-catalyzed biosynthesis of GAA is decreased. That high GAA/hArg is associated with a higher risk for all-cause and cardiovascular mortality may suggest that low plasma hArg is a stronger contributor to these adverse outcomes in RTR than GAA.
Collapse
Affiliation(s)
- Erik Hanff
- Core Unit Proteomics, Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Mohammad Yusof Said
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Arslan Arinc Kayacelebi
- Core Unit Proteomics, Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Adrian Post
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Isidor Minovic
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Else van den Berg
- Division of Acute Medicine, Department of Internal Medicine, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Martin H de Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Dimitrios Tsikas
- Core Unit Proteomics, Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
33
|
Van den Bergh G, Opdebeeck B, D'Haese PC, Verhulst A. The Vicious Cycle of Arterial Stiffness and Arterial Media Calcification. Trends Mol Med 2019; 25:1133-1146. [PMID: 31522956 DOI: 10.1016/j.molmed.2019.08.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/13/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
Arterial media calcification and arterial stiffness are independent predictors of cardiovascular mortality. Both processes reinforce one another, creating a vicious cycle in which transdifferentiation of endothelial cells and vascular smooth muscle cells play a central role. Physiological functioning of vascular smooth muscle cells in the arterial medial layer greatly depends on normal endothelial cell behavior. Endothelial or intimal layer cells are the primary sensors of pathological triggers circulating in the blood during, for example, ageing or inflammation, and often can be seen as initiators of this vicious cycle. As such, the search for treatment of arterial media calcification, which until now has been mainly concentrated at the level of the vascular smooth cell, may need to be expanded to intimal layer targets.
Collapse
Affiliation(s)
- Geoffrey Van den Bergh
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, B-2610 Wilrijk, Belgium
| | - Britt Opdebeeck
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, B-2610 Wilrijk, Belgium
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, B-2610 Wilrijk, Belgium.
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, B-2610 Wilrijk, Belgium
| |
Collapse
|
34
|
Inhibition of vascular smooth muscle cell calcification by vasorin through interference with TGFβ1 signaling. Cell Signal 2019; 64:109414. [PMID: 31505229 DOI: 10.1016/j.cellsig.2019.109414] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 01/05/2023]
Abstract
Elevated transforming growth factor β1 (TGFβ1) levels are frequently observed in chronic kidney disease (CKD) patients. TGFβ1 contributes to development of medial vascular calcification during hyperphosphatemia, a pathological process promoted by osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs). Vasorin is a transmembrane glycoprotein highly expressed in VSMCs, which is able to bind TGFβ to inhibit TGFβ signaling. Thus, the present study explored the effects of vasorin on osteo-/chondrogenic transdifferentiation and calcification of VSMCs. Primary human aortic smooth muscle cells (HAoSMCs) were treated with recombinant human TGFβ1 or β-glycerophosphate without or with recombinant human vasorin or vasorin gene silencing by siRNA. As a result, TGFβ1 down-regulated vasorin mRNA expression in HAoSMCs. Vasorin supplementation inhibited TGFβ1-induced pathway activation, SMAD2 phosphorylation and downstream target genes expression in HAoSMCs. Furthermore, treatment with exogenous vasorin blunted, while vasorin knockdown augmented TGFβ1-induced osteo-/chondrogenic transdifferentiation of HAoSMCs. In addition, phosphate down-regulated vasorin mRNA expression in HAoSMCs. Phosphate-induced TGFβ1 expression was not affected by addition of exogenous vasorin. Nonetheless, the phosphate-induced TGFβ1 signaling, osteo-/chondrogenic transdifferentiation and calcification of HAoSMCs were all blunted by vasorin. Conversely, silencing of vasorin aggravated osteoinduction in HAoSMCs during high phosphate conditions. Aortic vasorin expression was reduced in the hyperphosphatemic klotho-hypomorphic mouse model of CKD-related vascular calcification. In conclusion, vasorin, which suppresses TGFβ1 signaling and protects against osteo-/chondrogenic transdifferentiation and calcification of VSMCs, is reduced by pro-calcifying conditions. Thus, vasorin is a novel key regulator of VSMC calcification and may represent a potential therapeutic target for vascular calcification during CKD.
Collapse
|
35
|
Would acetazolamide inhibit progression of atheromatous vascular calcification? Med Hypotheses 2019; 132:109354. [PMID: 31421415 DOI: 10.1016/j.mehy.2019.109354] [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: 06/28/2019] [Revised: 07/25/2019] [Accepted: 08/05/2019] [Indexed: 10/26/2022]
Abstract
Vascular calcification is a recognised source of morbidity among mid-age and elderly subjects. Its development follows classical mineralisation pathways, inhibited by acidosis. It is known that the final mechanism of tissue mineralization involves three processes, all of which are highly pH dependent. Calcium interacts with phosphate in its trivalent form, but this step is inhibited by pyrophosphate, itself a source of phosphate when hydrolysed by alkaline phosphatase. Separately, matrix vesicles create nucleation sites and may indirectly disrupt vascular smooth muscle cells. Metabolic acidosis acts at every point to delay mineralization. The diuretic acetazolamide creates a sustained mild acidosis with some phosphate loss and, though usually ineffective in the experimental model, has been used with success in certain clinical conditions. We suggest that acetazolamide, well studied and tolerated, might inhibit progression of vascular calcification in subjects at risk through its dual action of lowering tissue pH and local phosphate concentration.
Collapse
|
36
|
Ma K, Liu P, Al-Maghout T, Sukkar B, Cao H, Voelkl J, Alesutan I, Pieske B, Lang F. Phosphate-induced ORAI1 expression and store-operated Ca 2+ entry in aortic smooth muscle cells. J Mol Med (Berl) 2019; 97:1465-1475. [PMID: 31385016 DOI: 10.1007/s00109-019-01824-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 07/02/2019] [Accepted: 07/25/2019] [Indexed: 12/16/2022]
Abstract
Compromised renal phosphate elimination in chronic kidney disease (CKD) leads to hyperphosphatemia, which in turn triggers osteo-/chondrogenic signaling in vascular smooth muscle cells (VSMCs) and vascular calcification. Osteo-/chondrogenic transdifferentiation of VSMCs leads to upregulation of the transcription factors MSX2, CBFA1, and SOX9 as well as tissue-nonspecific alkaline phosphatase (ALPL) which fosters calcification by degrading the calcification inhibitor pyrophosphate. Osteo-/chondrogenic signaling in VSMCs involves the serum- and glucocorticoid-inducible kinase SGK1. As shown in other cell types, SGK1 is a powerful stimulator of ORAI1, a Ca2+-channel accomplishing store-operated Ca2+-entry (SOCE). ORAI1 is stimulated following intracellular store depletion by the Ca2+ sensor STIM1. The present study explored whether phosphate regulates ORAI1 and/or STIM1 expression and, thus, SOCE in VSMCs. To this end, primary human aortic smooth muscle cells (HAoSMCs) were exposed to the phosphate donor β-glycerophosphate. Transcript levels were estimated by qRT-PCR, protein abundance by western blotting, ALPL activity by colorimetry, calcification by alizarin red S staining, 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. As a result, β-glycerophosphate treatment increased ORAI1 and STIM1 transcript levels and protein abundance as well as SOCE in HAoSMCs. Additional treatment with ORAI1 inhibitor MRS1845 or SGK1 inhibitor GSK650394 virtually disrupted the effects of β-glycerophosphate on SOCE. Moreover, the β-glycerophosphate-induced MSX2, CBFA1, SOX9, and ALPL mRNA expression and activity in HAoSMCs were suppressed in the presence of the ORAI1 inhibitor and upon ORAI1 silencing. In conclusion, enhanced phosphate upregulates ORAI1 and STIM1 expression and store-operated Ca2+-entry, which participate in the orchestration of osteo-/chondrogenic signaling of VSMCs. KEY MESSAGES: • In aortic SMC, phosphate donor ß-glycerophosphate upregulates Ca2+ channel ORAI1. • In aortic SMC, ß-glycerophosphate upregulates ORAI1-activator STIM1. • In aortic SMC, ß-glycerophosphate upregulates store-operated Ca2+-entry (SOCE). • The effect of ß-glycerophosphate on SOCE is disrupted by ORAI1 inhibitor MRS1845. • Stimulation of osteogenic signaling is disrupted by MRS1845 and ORAI1 silencing.
Collapse
Affiliation(s)
- Ke Ma
- Department of Pharmacology & Experimental Therapy, University of Tübingen, 72076, Tübingen, Germany
| | - Ping Liu
- Department of Pharmacology & Experimental Therapy, University of Tübingen, 72076, Tübingen, Germany
| | - Tamer Al-Maghout
- Department of Pharmacology & Experimental Therapy, University of Tübingen, 72076, Tübingen, Germany
| | - Basma Sukkar
- Department of Pharmacology & Experimental Therapy, University of Tübingen, 72076, Tübingen, Germany
| | - Hang Cao
- Department of Pharmacology & Experimental Therapy, University of Tübingen, 72076, Tübingen, Germany
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, 4040, Linz, Austria.,Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany.,Department of Nephrology and Medical Intensive Care, Charité University Medicine, Berlin, Germany
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, 4040, Linz, Austria.,Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Berlin, Germany
| | - Florian Lang
- Department of Vegetative and Clinical Physiology, University of Tübingen, Wilhelmstr. 56, 72074, Tübingen, Germany.
| |
Collapse
|
37
|
Henze LA, Luong TT, Boehme B, Masyout J, Schneider MP, Brachs S, Lang F, Pieske B, Pasch A, Eckardt KU, Voelkl J, Alesutan I. Impact of C-reactive protein on osteo-/chondrogenic transdifferentiation and calcification of vascular smooth muscle cells. Aging (Albany NY) 2019; 11:5445-5462. [PMID: 31377747 PMCID: PMC6710049 DOI: 10.18632/aging.102130] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 07/25/2019] [Indexed: 05/01/2023]
Abstract
Medial vascular calcification occurs during the aging process and is strongly accelerated by chronic kidney disease (CKD). Elevated C-reactive protein (CRP) levels are associated with vascular calcification, cardiovascular events and mortality in CKD patients. CRP is an important promoter of vascular inflammation. Inflammatory processes are critically involved in initiation and progression of vascular calcification. Thus, the present study explored a possible impact of CRP on vascular calcification. We found that CRP promoted osteo-/chondrogenic transdifferentiation and aggravated phosphate-induced osteo-/chondrogenic transdifferentiation and calcification of primary human aortic smooth muscle cells (HAoSMCs). These effects were paralleled by increased cellular oxidative stress and corresponding pro-calcific downstream-signaling. Antioxidants or p38 MAPK inhibition suppressed CRP-induced osteo-/chondrogenic signaling and mineralization. Furthermore, silencing of Fc fragment of IgG receptor IIa (FCGR2A) blunted the pro-calcific effects of CRP. Vascular CRP expression was increased in the klotho-hypomorphic mouse model of aging as well as in HAoSMCs during calcifying conditions. In conclusion, CRP augments osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells through mechanisms involving FCGR2A-dependent induction of oxidative stress. Thus, systemic inflammation may actively contribute to the progression of vascular calcification.
Collapse
MESH Headings
- Aging/metabolism
- Aging/pathology
- Animals
- C-Reactive Protein/metabolism
- Cell Transdifferentiation/physiology
- Cells, Cultured
- Chondrogenesis/physiology
- Disease Models, Animal
- Glucuronidase/genetics
- Glucuronidase/metabolism
- Humans
- Klotho Proteins
- Mice
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Osteogenesis/physiology
- Oxidative Stress
- RNA, Small Interfering/genetics
- Receptors, IgG/antagonists & inhibitors
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Signal Transduction
- Vascular Calcification/etiology
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- Laura A. Henze
- Department of Internal Medicine and Cardiology, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany
| | - Trang T.D. Luong
- Department of Internal Medicine and Cardiology, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz 4040, Austria
| | - Beate Boehme
- Department of Internal Medicine and Cardiology, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany
| | - Jaber Masyout
- Department of Internal Medicine and Cardiology, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany
| | - Markus P. Schneider
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Sebastian Brachs
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin 10115, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin 10115, Germany
| | - Florian Lang
- Department of Physiology I, Eberhard-Karls University, Tübingen 72076, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin 10115, Germany
- Berlin Institute of Health (BIH), Berlin 10178, Germany
- Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Berlin 13353, Germany
| | - Andreas Pasch
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz 4040, Austria
- Calciscon AG, 2560 Nidau-Biel, Switzerland
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Jakob Voelkl
- Department of Internal Medicine and Cardiology, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz 4040, Austria
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin 10115, Germany
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Ioana Alesutan
- Department of Internal Medicine and Cardiology, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz 4040, Austria
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin 10115, Germany
- Berlin Institute of Health (BIH), Berlin 10178, Germany
| |
Collapse
|
38
|
Rodionov RN, Begmatov H, Jarzebska N, Patel K, Mills MT, Ghani Z, Khakshour D, Tamboli P, Patel MN, Abdalla M, Assaf M, Bornstein SR, Millan JL, Bode-Böger SM, Martens-Lobenhoffer J, Weiss N, Savinova OV. Homoarginine Supplementation Prevents Left Ventricular Dilatation and Preserves Systolic Function in a Model of Coronary Artery Disease. J Am Heart Assoc 2019; 8:e012486. [PMID: 31304837 PMCID: PMC6662144 DOI: 10.1161/jaha.119.012486] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background Homoarginine (hArg) has been shown to be cardioprotective in a model of ischemic heart failure; however, the mechanism remains unknown. hArg can inhibit tissue‐nonspecific alkaline phosphatase (TNAP), an enzyme that promotes vascular calcification. We hypothesized that hArg will exert beneficial effects by reducing calcification in a mouse model of coronary artery disease associated with TNAP overexpression and hypercholesterolemia. Methods and Results TNAP was overexpressed in the endothelium in mice homozygous for a low‐density lipoprotein receptor mutation (wicked high cholesterol [WHC] allele). WHC and WHC–endothelial TNAP mice received placebo or hArg supplementation (14 mg/L in drinking water) starting at 6 weeks of age simultaneously with an atherogenic diet. Outcomes were compared between the groups after 4 to 5 weeks on treatment. Experiments were performed in males, which presented a study limitation. As expected, WHC–endothelial TNAP mice on the placebo had increased mortality (median survival 27 days, P<0.0001), increased coronary calcium and lipids (P<0.01), increased left ventricular end‐diastolic diameter (P<0.0001), reduced ejection fraction (P<0.05), and increased myocardial fibrosis (P<0.0001) compared with WHC mice. Contrary to our hypothesis, hArg neither inhibited TNAP activity in vivo nor reduced coronary artery calcification and atherosclerosis in WHC–endothelial TNAP mice; however, compared with the placebo, hArg prevented left ventricular dilatation (P<0.01), preserved ejection fraction (P<0.05), and reduced myocardial fibrosis (P<0.001). Conclusions The beneficial effect of hArg supplementation in the setting of calcified coronary artery disease is likely due to its direct protective actions on the myocardial response to the ischemic injury and not to the inhibition of TNAP activity and calcification.
Collapse
Affiliation(s)
- Roman N Rodionov
- 1 University Center for Vascular Medicine Technische Universität Dresden Dresden Germany
| | - Hoshimjon Begmatov
- 2 Department of Biomedical Sciences New York Institute of Technology College of Osteopathic Medicine Old Westbury NY
| | - Natalia Jarzebska
- 1 University Center for Vascular Medicine Technische Universität Dresden Dresden Germany.,3 Department of Anesthesiology and Intensive Care Unit Medicine Pulmonary Engineering Group University Hospital Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Ketul Patel
- 2 Department of Biomedical Sciences New York Institute of Technology College of Osteopathic Medicine Old Westbury NY
| | - Matthew T Mills
- 2 Department of Biomedical Sciences New York Institute of Technology College of Osteopathic Medicine Old Westbury NY
| | - Zulaikha Ghani
- 2 Department of Biomedical Sciences New York Institute of Technology College of Osteopathic Medicine Old Westbury NY
| | - Doreen Khakshour
- 2 Department of Biomedical Sciences New York Institute of Technology College of Osteopathic Medicine Old Westbury NY
| | - Pankti Tamboli
- 2 Department of Biomedical Sciences New York Institute of Technology College of Osteopathic Medicine Old Westbury NY
| | - Mitul N Patel
- 2 Department of Biomedical Sciences New York Institute of Technology College of Osteopathic Medicine Old Westbury NY
| | - Mirette Abdalla
- 2 Department of Biomedical Sciences New York Institute of Technology College of Osteopathic Medicine Old Westbury NY
| | - Maryann Assaf
- 2 Department of Biomedical Sciences New York Institute of Technology College of Osteopathic Medicine Old Westbury NY
| | - Stefan R Bornstein
- 4 Department of Internal Medicine III University Hospital Carl Gustav Carus Technische Universität Dresden Dresden Germany
| | - Jose Luis Millan
- 5 Human Genetics Program Sanford Burnham Prebys Medical Discovery Institute La Jolla CA
| | | | | | - Norbert Weiss
- 1 University Center for Vascular Medicine Technische Universität Dresden Dresden Germany
| | - Olga V Savinova
- 2 Department of Biomedical Sciences New York Institute of Technology College of Osteopathic Medicine Old Westbury NY
| |
Collapse
|
39
|
Abstract
Vascular calcification results from an imbalance of promoters and inhibitors of mineralization in the vascular wall, culminating in the creation of an organized extracellular matrix deposition. It is characterized by the accumulation of calcium phosphate complex and crystallization of hydroxyapatite in the tunica media, leading to vessel stiffening. The underlying initiators of dysregulated calcification maintenance are diverse. These range from the expression of bone-associated proteins, to the osteogenic transdifferentiation of smooth muscle cells to osteoblast-like cells, to the release of fragmented apoptotic bodies and mineralization competent extracellular vesicles by smooth muscle cells, which act as a nucleation site for the deposition of hydroxyapatite crystals. The process involves a complex interplay between vitamin K-dependent calcification-inhibitory proteins, such as matrix γ-carboxyglutamate acid (Gla) protein, Gla-rich protein and growth arrest-specific gene 6 protein, and stimulatory mediators, such as osteocalcin. Vitamin K plays an important role as a cofactor for posttranslational γ-carboxylation of matrix Gla proteins in converting to a biologically active conformation. Drugs that inhibit vitamin K, such as warfarin, impair γ-carboxylation of Gla proteins, resulting in the accumulation of uncarboxylated proteins lacking calcification-inhibitory capacity. This article overviews the involvement of systemically and locally expressed vitamin K-dependent proteins in vascular calcification and their potential as biomarkers of calcification.
Collapse
Affiliation(s)
- Belay Tesfamariam
- 1 Division of Cardiovascular and Renal Products, Center for Drug Evaluation and Research, FDA, Silver Spring, MD, USA
| |
Collapse
|
40
|
Bollenbach A, Cordts K, Hanff E, Atzler D, Choe CU, Schwedhelm E, Tsikas D. Evidence by GC-MS that lysine is an arginase-catalyzed metabolite of homoarginine in vitro and in vivo in humans. Anal Biochem 2019; 577:59-66. [DOI: 10.1016/j.ab.2019.04.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/23/2019] [Accepted: 04/23/2019] [Indexed: 10/27/2022]
|
41
|
Voelkl J, Lang F, Eckardt KU, Amann K, Kuro-O M, Pasch A, Pieske B, Alesutan I. Signaling pathways involved in vascular smooth muscle cell calcification during hyperphosphatemia. Cell Mol Life Sci 2019; 76:2077-2091. [PMID: 30887097 PMCID: PMC6502780 DOI: 10.1007/s00018-019-03054-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 02/06/2023]
Abstract
Medial vascular calcification has emerged as a putative key factor contributing to the excessive cardiovascular mortality of patients with chronic kidney disease (CKD). Hyperphosphatemia is considered a decisive determinant of vascular calcification in CKD. A critical role in initiation and progression of vascular calcification during elevated phosphate conditions is attributed to vascular smooth muscle cells (VSMCs), which are able to change their phenotype into osteo-/chondroblasts-like cells. These transdifferentiated VSMCs actively promote calcification in the medial layer of the arteries by producing a local pro-calcifying environment as well as nidus sites for precipitation of calcium and phosphate and growth of calcium phosphate crystals. Elevated extracellular phosphate induces osteo-/chondrogenic transdifferentiation of VSMCs through complex intracellular signaling pathways, which are still incompletely understood. The present review addresses critical intracellular pathways controlling osteo-/chondrogenic transdifferentiation of VSMCs and, thus, vascular calcification during hyperphosphatemia. Elucidating these pathways holds a significant promise to open novel therapeutic opportunities counteracting the progression of vascular calcification in CKD.
Collapse
MESH Headings
- Animals
- Calcium Phosphates/chemistry
- Calcium Phosphates/metabolism
- Cell Transdifferentiation
- Chondrocytes/metabolism
- Chondrocytes/pathology
- Gene Expression Regulation
- Humans
- Hyperphosphatemia/complications
- Hyperphosphatemia/genetics
- Hyperphosphatemia/metabolism
- Hyperphosphatemia/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Osteoblasts/metabolism
- Osteoblasts/pathology
- RANK Ligand/genetics
- RANK Ligand/metabolism
- Receptor Activator of Nuclear Factor-kappa B/genetics
- Receptor Activator of Nuclear Factor-kappa B/metabolism
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Signal Transduction
- Vascular Calcification/complications
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040, Linz, Austria.
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany.
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353, Berlin, Germany.
| | - Florian Lang
- Department of Physiology I, Eberhard-Karls University, Wilhelmstr. 56, 72076, Tübingen, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353, Berlin, Germany
| | - Kerstin Amann
- Department of Nephropathology, Universität Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Andreas Pasch
- Calciscon AG, Aarbergstrasse 5, 2560, Nidau-Biel, Switzerland
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch Str. 2, 10178, Berlin, Germany
- Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040, Linz, Austria
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch Str. 2, 10178, Berlin, Germany
| |
Collapse
|
42
|
Schanstra JP, Luong TT, Makridakis M, Van Linthout S, Lygirou V, Latosinska A, Alesutan I, Boehme B, Schelski N, Von Lewinski D, Mullen W, Nicklin S, Delles C, Feuillet G, Denis C, Lang F, Pieske B, Bascands JL, Mischak H, Saulnier-Blache JS, Voelkl J, Vlahou A, Klein J. Systems biology identifies cytosolic PLA2 as a target in vascular calcification treatment. JCI Insight 2019; 4:125638. [PMID: 31092728 DOI: 10.1172/jci.insight.125638] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/17/2019] [Indexed: 01/15/2023] Open
Abstract
Although cardiovascular disease (CVD) is the leading cause of morbimortality worldwide, promising new drug candidates are lacking. We compared the arterial high-resolution proteome of patients with advanced versus early-stage CVD to predict, from a library of small bioactive molecules, drug candidates able to reverse this disease signature. Of the approximately 4000 identified proteins, 100 proteins were upregulated and 52 were downregulated in advanced-stage CVD. Arachidonyl trifluoromethyl ketone (AACOCF3), a cytosolic phospholipase A2 (cPLA2) inhibitor was predicted as the top drug able to reverse the advanced-stage CVD signature. Vascular cPLA2 expression was increased in patients with advanced-stage CVD. Treatment with AACOCF3 significantly reduced vascular calcification in a cholecalciferol-overload mouse model and inhibited osteoinductive signaling in vivo and in vitro in human aortic smooth muscle cells. In conclusion, using a systems biology approach, we have identified a potentially new compound that prevented typical vascular calcification in CVD in vivo. Apart from the clear effect of this approach in CVD, such strategy should also be able to generate novel drug candidates in other complex diseases.
Collapse
Affiliation(s)
- Joost P Schanstra
- Institute of Cardiovascular and Metabolic Disease, INSERM, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Trang Td Luong
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Manousos Makridakis
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Sophie Van Linthout
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies (BCRT), Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Vasiliki Lygirou
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Ioana Alesutan
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Beate Boehme
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Nadeshda Schelski
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | | | - William Mullen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Stuart Nicklin
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Guylène Feuillet
- Institute of Cardiovascular and Metabolic Disease, INSERM, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Colette Denis
- Institute of Cardiovascular and Metabolic Disease, INSERM, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Florian Lang
- Department of Physiology I, University of Tubingen, Tubingen, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Jean-Loup Bascands
- INSERM, U1188, Université de La Réunion, Sainte-Clotilde, La Réunion, France
| | | | - Jean-Sebastien Saulnier-Blache
- Institute of Cardiovascular and Metabolic Disease, INSERM, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Jakob Voelkl
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany.,Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Antonia Vlahou
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Julie Klein
- Institute of Cardiovascular and Metabolic Disease, INSERM, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| |
Collapse
|
43
|
Asymmetric dimethylation and citrullination of proteinic arginine and homoarginine synthesis in human Helicobacter pylori infection. Amino Acids 2019; 51:961-971. [DOI: 10.1007/s00726-019-02737-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/12/2019] [Indexed: 12/20/2022]
|
44
|
Babic M, Schuchardt M, Tölle M, van der Giet M. In times of tobacco-free nicotine consumption: The influence of nicotine on vascular calcification. Eur J Clin Invest 2019; 49:e13077. [PMID: 30721530 DOI: 10.1111/eci.13077] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 01/11/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Smoking remains the most important avoidable cause of global mortality. Even though the number of cigarette smokers declines in first world countries, the uses of alternative nicotine delivery products increase and may even surpass the sells of cigarettes. In this light, the explicit role of nicotine in the development of cardiovascular diseases should be elucidated. OBJECTIVES This narrative review attempts to connect current literature about possible effects of nicotine on the environment of the vasculature to the pathogenesis of vascular calcification, focusing on the tunica media of the vessel wall. METHODS For this review, papers found on Pubmed and Medline until December 2018 by searching for the keywords nicotine, vascular calcification, oxidative stress, osteoblastic transdifferentiation and matrix degradation were considered. RESULTS Nicotine creates an environment that probably facilitates and maybe even induces osteogenic transdifferentiation of VSMC by inflammation, endothelial dysfunction and reactive oxygen species. This process is believed to be a key event in calcification of the tunica media of the vessel wall. Furthermore, nicotine could lead to the formation of nucleation sites for hydroxyapatite by facilitating matrix vesicles and extracellular matrix degradation. CONCLUSIONS There is a growing body of evidence implicating that nicotine alone could impair vascular function and lead to vascular calcification. Further research is necessary to elucidate the explicit influence of nicotine on arteriosclerosis.
Collapse
Affiliation(s)
- Milen Babic
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Mirjam Schuchardt
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus Tölle
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Markus van der Giet
- Department of Nephrology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
45
|
Nitz K, Lacy M, Atzler D. Amino Acids and Their Metabolism in Atherosclerosis. Arterioscler Thromb Vasc Biol 2019; 39:319-330. [DOI: 10.1161/atvbaha.118.311572] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
As a leading cause of death worldwide, cardiovascular disease is a global health concern. The development and progression of atherosclerosis, which ultimately gives rise to cardiovascular disease, has been causally linked to hypercholesterolemia. Mechanistically, the interplay between lipids and the immune system during plaque progression significantly contributes to the chronic inflammation seen in the arterial wall during atherosclerosis. Localized inflammation and increased cell-to-cell interactions may influence polarization and proliferation of immune cells via changes in amino acid metabolism. Specifically, the amino acids
l
-arginine (Arg),
l
-homoarginine (hArg) and
l
-tryptophan (Trp) have been widely studied in the context of cardiovascular disease, and their metabolism has been established as key regulators of vascular homeostasis, as well as immune cell function. Cyclic effects between endothelial cells, innate, and adaptive immune cells exist during Arg and hArg, as well as Trp metabolism, that may have distinct effects on the development of atherosclerosis. In this review, we describe the current knowledge surrounding the metabolism, biological function, and clinical perspective of Arg, hArg, and Trp in the context of atherosclerosis.
Collapse
Affiliation(s)
- Katrin Nitz
- From the Institute for Cardiovascular Prevention (K.N., M.L., D.A.), Ludwig-Maximilians-University, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.N., M.L., D.A.)
| | - Michael Lacy
- From the Institute for Cardiovascular Prevention (K.N., M.L., D.A.), Ludwig-Maximilians-University, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.N., M.L., D.A.)
| | - Dorothee Atzler
- From the Institute for Cardiovascular Prevention (K.N., M.L., D.A.), Ludwig-Maximilians-University, Munich, Germany
- Walther Straub Institute of Pharmacology and Toxicology (D.A.), Ludwig-Maximilians-University, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.N., M.L., D.A.)
| |
Collapse
|
46
|
SGK1-dependent stimulation of vascular smooth muscle cell osteo-/chondrogenic transdifferentiation by interleukin-18. Pflugers Arch 2019; 471:889-899. [PMID: 30706178 PMCID: PMC6533237 DOI: 10.1007/s00424-019-02256-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/04/2019] [Accepted: 01/13/2019] [Indexed: 01/18/2023]
Abstract
The serum- and glucocorticoid-inducible kinase 1 (SGK1) is a key regulator of osteo-/chondrogenic transdifferentiation and subsequent calcification of vascular smooth muscle cells (VSMCs). The phenotypical transdifferentiation of VSMCs is associated with increased interleukin-18 (IL-18) levels and generalized inflammation. Therefore, the present study investigated the possible involvement of SGK1 in IL-18-induced vascular calcification. Experiments were performed in primary human aortic smooth muscle cells (HAoSMCs) treated with recombinant human IL-18 protein in control or high phosphate conditions and following SGK1 knockdown by siRNA or pharmacological inhibition of SGK1, PI3K, and PDK1. As a result, IL-18 treatment increased SGK1 mRNA and protein expression in HAoSMCs. IL-18 upregulated SGK1 mRNA expression in a dose-dependent manner. This effect was paralleled by upregulation of the mRNA expression of MSX2 and CBFA1, osteogenic transcription factors, and of tissue-nonspecific alkaline phosphatase (ALPL), an osteogenic enzyme, as markers of increased osteo-/chondrogenic transdifferentiation. Phosphate treatment increased SGK1 and osteogenic markers mRNA expression as well as ALPL activity and induced calcification of HAoSMCs, all effects significantly augmented by additional treatment with IL-18. Conversely, silencing of SGK1 or cotreatment with the SGK1 inhibitor EMD638683 blunted the effects of IL-18 on osteo-/chondrogenic transdifferentiation and calcification of HAoSMCs. The procalcific effects of IL-18 were similarly suppressed in the presence of PI3K or PDK1 inhibitors. In conclusion, SGK1 expression is upregulated by IL-18 in VSMCs and SGK1 participates in the intracellular signaling of IL-18-induced osteo-/chondrogenic transdifferentiation of VSMCs. Thus, SGK1 may serve as therapeutic target to limit the progression of medial vascular calcification during vascular inflammation.
Collapse
|
47
|
Hanff E, Ruben S, Kreuzer M, Bollenbach A, Kayacelebi AA, Das AM, von Versen-Höynck F, von Kaisenberg C, Haffner D, Ückert S, Tsikas D. Development and validation of GC–MS methods for the comprehensive analysis of amino acids in plasma and urine and applications to the HELLP syndrome and pediatric kidney transplantation: evidence of altered methylation, transamidination, and arginase activity. Amino Acids 2019; 51:529-547. [DOI: 10.1007/s00726-018-02688-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/12/2018] [Indexed: 12/19/2022]
|
48
|
Affiliation(s)
- Heather Y Small
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, UK
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, UK.,Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, 31-008 Anny 12, Krakow, Poland
| |
Collapse
|
49
|
Plasma Homoarginine Concentrations According to Use of Hormonal Contraception. Sci Rep 2018; 8:12217. [PMID: 30111829 PMCID: PMC6093891 DOI: 10.1038/s41598-018-30708-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 07/30/2018] [Indexed: 11/09/2022] Open
Abstract
Estrogen is a potent vasodilator through activation of endothelial nitric oxide synthase (eNOS). Arginine and its homologue homoarginine are substrates for NOS, while asymmetric dimethylarginine (ADMA) is a NOS inhibitor. Healthy, never-pregnant women aged 18 to 40 years (n = 158) were categorized according to use of hormonal contraception into non-users (n = 76), users of estrogen contraceptives (EC-users, n = 58) and users of progestins-only contraceptives (PC-users, n = 24). Plasma homoarginine, arginine, ADMA and SDMA concentrations were assayed using a LC-MS/MS method. Compared to non-users, EC users had higher plasma homoarginine (median (interquartile range) 1.63 (1.24, 2.04) vs. 2.39 (2.05, 2.85) µmol/L, p < 0.001), lower arginine (80.8 (72.4, 94.3) vs. 72.1 (62.9, 85.1) µmol/L, p = 0.008) and ADMA (0.52 (0.46, 0.59) vs. 0.48 (0.42, 0.54) µmol/L, p = 0.003) concentrations. The lowest median plasma homoarginine concentration (1.34 (0.92, 1.75) µmol) was seen in PC-users. No differences were seen in SDMA concentrations according to use of hormonal contraception. In healthy, never-pregnant women aged 18 to 40 years, use of estrogen containing contraception was associated with significantly higher plasma concentrations of homoarginine and lower plasma concentrations of arginine and ADMA as compared to non-users, while the lowest plasma homoarginine concentrations were seen in progestin-only users. Whether the observed changes in relation to use of hormonal contraception have an impact on cardiovascular status, should be evaluated in an intervention study.
Collapse
|
50
|
Tuffaha R, Voelkl J, Pieske B, Lang F, Alesutan I. Role of PKB/SGK-dependent phosphorylation of GSK-3α/β in vascular calcification during cholecalciferol overload in mice. Biochem Biophys Res Commun 2018; 503:2068-2074. [PMID: 30119888 DOI: 10.1016/j.bbrc.2018.07.161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022]
Abstract
Medial vascular calcification is a highly regulated process involving osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells. Both, protein kinase B (PKB) and serum- and glucocorticoid-inducible kinase 1 (SGK1) are involved in the intracellular signaling of vascular calcification and both phosphorylate and inactivate glycogen synthase kinase 3 (GSK-3). The present study explored whether PKB/SGK-dependent phosphorylation of GSK-3α/β is involved in vascular calcification. Experiments were performed in Gsk-3α/β double knockin mice lacking functional PKB/SGK phosphorylation sites (gsk-3KI) and corresponding wild-type mice (gsk-3WT) following high-dosed cholecalciferol treatment as well as ex vivo in aortic ring explants from gsk-3KI and gsk-3WT mice treated without and with phosphate. In gsk-3WT mice, high-dosed cholecalciferol induced vascular calcification and aortic osteo-/chondrogenic signaling, shown by increased expression of osteogenic markers Msx2, Cbfa1 and tissue-nonspecific alkaline phosphatase (Alpl). All these effects were suppressed in aortic tissue from gsk-3KI mice. Cholecalciferol decreased aortic Gsk-3α/β phosphorylation (Ser21/9) in gsk-3WT mice, while no phosphorylation was observed in gsk-3KI mice. Moreover, the mRNA expression of type III sodium-dependent phosphate transporter (Pit1) and plasminogen activator inhibitor 1 (Pai1) was increased following cholecalciferol treatment in aortic tissue of gsk-3WT mice, effects again blunted in gsk-3KI mice. In addition, phosphate treatment induced mineral deposition and osteogenic markers expression in aortic ring explants from gsk-3WT mice, effects reduced in aortic ring explants from gsk-3KI mice. In conclusion, vascular PKB/SGK-dependent phosphorylation of GSK-3α/β contributes to the osteoinductive signaling leading to vascular calcification.
Collapse
Affiliation(s)
- Rashad Tuffaha
- Department of Physiology I, Eberhard-Karls University, Wilhelmstr. 56, 72076 Tübingen, Germany
| | - Jakob Voelkl
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Hessische Str. 3-4, 10115 Berlin, Germany.
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Hessische Str. 3-4, 10115 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Str. 2, 10178 Berlin, Germany; Department of Internal Medicine and Cardiology, German Heart Center Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Florian Lang
- Department of Physiology I, Eberhard-Karls University, Wilhelmstr. 56, 72076 Tübingen, Germany
| | - Ioana Alesutan
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Hessische Str. 3-4, 10115 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Str. 2, 10178 Berlin, Germany
| |
Collapse
|