1
|
Xue C, Chu Q, Shi Q, Zeng Y, Lu J, Li L. Wnt signaling pathways in biology and disease: mechanisms and therapeutic advances. Signal Transduct Target Ther 2025; 10:106. [PMID: 40180907 PMCID: PMC11968978 DOI: 10.1038/s41392-025-02142-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/13/2024] [Accepted: 12/29/2024] [Indexed: 04/05/2025] Open
Abstract
The Wnt signaling pathway is critically involved in orchestrating cellular functions such as proliferation, migration, survival, and cell fate determination during development. Given its pivotal role in cellular communication, aberrant Wnt signaling has been extensively linked to the pathogenesis of various diseases. This review offers an in-depth analysis of the Wnt pathway, detailing its signal transduction mechanisms and principal components. Furthermore, the complex network of interactions between Wnt cascades and other key signaling pathways, such as Notch, Hedgehog, TGF-β, FGF, and NF-κB, is explored. Genetic mutations affecting the Wnt pathway play a pivotal role in disease progression, with particular emphasis on Wnt signaling's involvement in cancer stem cell biology and the tumor microenvironment. Additionally, this review underscores the diverse mechanisms through which Wnt signaling contributes to diseases such as cardiovascular conditions, neurodegenerative disorders, metabolic syndromes, autoimmune diseases, and cancer. Finally, a comprehensive overview of the therapeutic progress targeting Wnt signaling was given, and the latest progress in disease treatment targeting key components of the Wnt signaling pathway was summarized in detail, including Wnt ligands/receptors, β-catenin destruction complexes, and β-catenin/TCF transcription complexes. The development of small molecule inhibitors, monoclonal antibodies, and combination therapy strategies was emphasized, while the current potential therapeutic challenges were summarized. This aims to enhance the current understanding of this key pathway.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
2
|
Zheng Y, Shao M, Zheng Y, Sun W, Qin S, Sun Z, Zhu L, Guan Y, Wang Q, Wang Y, Li L. PPARs in atherosclerosis: The spatial and temporal features from mechanism to druggable targets. J Adv Res 2025; 69:225-244. [PMID: 38555000 PMCID: PMC11954843 DOI: 10.1016/j.jare.2024.03.020] [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: 01/17/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Atherosclerosis is a chronic and complex disease caused by lipid disorder, inflammation, and other factors. It is closely related to cardiovascular diseases, the chief cause of death globally. Peroxisome proliferator-activated receptors (PPARs) are valuable anti-atherosclerosis targets that showcase multiple roles at different pathological stages of atherosclerosis and for cell types at different tissue sites. AIM OF REVIEW Considering the spatial and temporal characteristics of the pathological evolution of atherosclerosis, the roles and pharmacological and clinical studies of PPARs were summarized systematically and updated under different pathological stages and in different vascular cells of atherosclerosis. Moreover, selective PPAR modulators and PPAR-pan agonists can exert their synergistic effects meanwhile reducing the side effects, thereby providing novel insight into future drug development for precise spatial-temporal therapeutic strategy of anti-atherosclerosis targeting PPARs. KEY SCIENTIFIC Concepts of Review: Based on the spatial and temporal characteristics of atherosclerosis, we have proposed the importance of stage- and cell type-dependent precision therapy. Initially, PPARs improve endothelial cells' dysfunction by inhibiting inflammation and oxidative stress and then regulate macrophages' lipid metabolism and polarization to improve fatty streak. Finally, PPARs reduce fibrous cap formation by suppressing the proliferation and migration of vascular smooth muscle cells (VSMCs). Therefore, research on the cell type-specific mechanisms of PPARs can provide the foundation for space-time drug treatment. Moreover, pharmacological studies have demonstrated that several drugs or compounds can exert their effects by the activation of PPARs. Selective PPAR modulators (that specifically activate gene subsets of PPARs) can exert tissue and cell-specific effects. Furthermore, the dual- or pan-PPAR agonist could perform a better role in balancing efficacy and side effects. Therefore, research on cells/tissue-specific activation of PPARs and PPAR-pan agonists can provide the basis for precision therapy and drug development of PPARs.
Collapse
Affiliation(s)
- Yi Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingyan Shao
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yanfei Zheng
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenlong Sun
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Si Qin
- Lab of Food Function and Nutrigenomics, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Ziwei Sun
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Linghui Zhu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuanyuan Guan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qi Wang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China.
| | - Lingru Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
3
|
Zhang L, Sun Y, Lin Y, Li H, Huang Y, Tang N, Zhang X, Lu Y, Kovalev VA, Snezhko EV, Luo Y, Wang B. Cell calcification reverses the chemoresistance of cancer cells via the conversion of glycolipid metabolism. Biomaterials 2025; 314:122886. [PMID: 39427430 DOI: 10.1016/j.biomaterials.2024.122886] [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: 06/09/2024] [Revised: 09/19/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Drug resistance is an inherent challenge during cancer chemotherapy. Cancer cells favor fatty acid metabolism through metabolic reprogramming to achieve therapeutic resistance. However, an effective approach to overcoming the switch from glycolysis-dependent to fatty acid beta-oxidation-dependent anabolic and energy metabolism remains elusive. Here, we developed a macromolecular drug (folate-polySia, FpSA) to induce the extracellular microcalcification of cervical cancer cells with cisplatin resistance. Microcalcification attenuated the uptake of fatty acids and the beta-oxidation of fatty acids by mitochondrial dysfunction but boosted the glycolysis pathway. Consequently, cotreatment with Pt and FpSA inhibited cisplatin-resistant tumor growth and improved tumor-bearing mice's survival rates, indicating that FpSA switched fatty acid metabolism to glycolysis to sensitize cisplatin-resistant cells further. Taken together, cancer cell calcification induced by FpSA provides a reprogramming metabolic strategy for the treatment of chemotherapy-resistant tumors.
Collapse
Affiliation(s)
- Lihong Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yandi Sun
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Yindan Lin
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanhui Li
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Yuqiao Huang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Ning Tang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Xueyun Zhang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yin Lu
- College of Biological and Environmental Engineering, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Vassili A Kovalev
- Biomedical Image Analysis Department, The United Institute of Informatics Problems, National Academy of Sciences of Belarus, Minsk, 220012, Belarus
| | - Eduard V Snezhko
- Biomedical Image Analysis Department, The United Institute of Informatics Problems, National Academy of Sciences of Belarus, Minsk, 220012, Belarus
| | - Yan Luo
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Ben Wang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China; State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China; Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Hangzhou, 310029, China; Cancer Center, Zhejiang University, Hangzhou, 310029, China.
| |
Collapse
|
4
|
Zeng Z, Zhao Z, Yuan Q, Yang S, Wang Z, Wang Z, Zeng S, Li A, Chen Q, Zhu G, Xiao X, Luo G, Luo H, Li J, Zu X, Xie H, Liu J. Hepatic Steatosis Aggravates Vascular Calcification via Extracellular Vesicle-Mediated Osteochondrogenic Switch of Vascular Smooth Muscle Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408660. [PMID: 39680681 PMCID: PMC11791995 DOI: 10.1002/advs.202408660] [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] [Received: 07/26/2024] [Revised: 10/17/2024] [Indexed: 12/18/2024]
Abstract
The global incidence of metabolic dysfunction-associated fatty liver disease (MAFLD) has risen sharply. This condition is strongly associated with the risk of cardiovascular disease (CVD), but how MAFLD affects the development and progression of CVD, particularly concerning vascular calcification, remains unclear. Herein, extracellular vesicles (EVs) are identified from steatotic hepatocytes as a trigger that accelerated the progression of both vascular intimal and medial calcification. Steatotic hepatocytes are found to release more EVs, which are able to reach the vascular tissue, be taken up by vascular smooth muscle cells (VSMCs), and promote their osteogenic differentiation. Within these toxic vesicles, a protein cargo is identified called lectin galactoside-binding soluble 3 binding protein (Lgals3bp) that acted as a potent inducer of osteochondrogenic transformation in VSMCs. Both the inhibition of EV release and the liver-specific knockdown of Lgals3bp profoundly attenuated vascular calcification. This work partially explains the reason for the high incidence of vascular calcification in MAFLD and unveils a novel mechanism that may be used to prevent or treat cardiovascular complications in patients with MAFLD.
Collapse
Affiliation(s)
- Zhao‐Lin Zeng
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Institute of Cardiovascular DiseaseKey Lab for Arteriosclerology of Hunan ProvinceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Zhi‐Bo Zhao
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Qing Yuan
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Shi‐Qi Yang
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Zhen‐Xing Wang
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zuo Wang
- Institute of Cardiovascular DiseaseKey Lab for Arteriosclerology of Hunan ProvinceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Shi‐Yu Zeng
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - An‐Qi Li
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Qian Chen
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Guo‐Qiang Zhu
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Xin‐Hua Xiao
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Guang‐Hua Luo
- Department of RadiologyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Hai‐Yan Luo
- Department of GastroenterologyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Jiao‐Yang Li
- Department of Occupational and Environmental HealthSchool of Public HealthWuhan UniversityWuhan430071P. R. China
| | - Xu‐Yu Zu
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Hui Xie
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jiang‐Hua Liu
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| |
Collapse
|
5
|
Yu XH, Guo XN, Li K, Li JW, Wang K, Wang D, Liu BC. The Role of Wnt5a in Inflammatory Diseases. Immunology 2025; 174:203-212. [PMID: 39668514 DOI: 10.1111/imm.13882] [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: 06/22/2024] [Revised: 10/08/2024] [Accepted: 11/22/2024] [Indexed: 12/14/2024] Open
Abstract
Wnt5a plays an important role in cell development and maturation and is closely associated with various diseases, such as malignant tumours, metabolic disorders, fibrosis, growth and development. Recent studies have shown that Wnt5a expression and signal transduction are strongly involved in the inflammatory response. This study comprehensively reviewed the latest research progress on the association between Wnt5a and several inflammatory diseases, such as sepsis, asthma, chronic obstructive pulmonary disease, tuberculosis, rheumatoid arthritis, atherosclerosis and psoriasis vulgare. We elucidated the mechanism by which the Wnt5a protein is involved in the pathogenesis of these diseases, providing a basis for the prevention and treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Xin-Hua Yu
- Department of Pediatrics, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Xin-Ning Guo
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kui Li
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Jia-Wei Li
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Kaijin Wang
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bi-Cui Liu
- Department of Respiratory and Critical Care Medicine, Bishan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Qi RQ, Chen YF, Cheng J, Song JW, Chen YH, Wang SY, Liu Y, Yan KX, Liu XY, Li J, Zhong JC. Elabela alleviates cuproptosis and vascular calcification in vitaminD3- overloaded mice via regulation of the PPAR-γ /FDX1 signaling. Mol Med 2024; 30:223. [PMID: 39567863 PMCID: PMC11577739 DOI: 10.1186/s10020-024-00997-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Vascular calcification is a crucial pathophysiological process associated with age-related cardiovascular diseases. Elabela, a recently identified peptide, has emerged as a significant player in the regulation of cardiovascular function and homeostasis. However, the effects and underlying mechanisms of Elabela on age-related vascular calcification remain largely unexplored. METHODS In-vivo vascular calcifications of C57BL/6J mice (8-week-old) and young (8-week-old) or aged (72-week-old) SD rats were injected with vitamin D3 (VitD3) or saline, respectively. Furthermore, the VitD3-overloaded mice received Elabela (1 mg/kg/d), peroxisome proliferators-activated receptor-γ (PPAR-γ) activator Rosiglitazone (5 mg/kg/d) or copper-ionophore Elesclomol (20 mg/kg/d), respectively. As for in-vitro studies, primary rat vascular smooth muscle cells (VSMCs) were isolated from aortas and cultured for explore the role and underlying mechanism of Elabela in vascular calcification. RESULTS There were marked increases in FDX1 and Slc31a1 levels in both aortas and VSMCs during vascular calcification, coinciding with a rise in copper levels and a decrease in Elabela levels. Alizarin red and von-Kossa staining indicated that the administration of Elabela effectively hindered the progression of vascular cuproptosis and arterial calcification in VitD3-overloaded mice and rat arterial rings models. Moreover, Elabela significantly suppressed osteogenic differentiation and calcium deposition in VSMCs and strikingly reversed high phosphate-induced augmentation of FDX1 expression, DLAT aggregation as well as intracellular copper ion levels. More importantly, Elabela exhibited remarkable abilities to prevent mitochondrial dysfunctions in primary rat VSMCs by maintaining mitochondrial membrane potential, inhibiting mitochondrial division, reducing mitochondrial ROS production and increasing ATP levels. Interestingly, Elabela mitigated cellular senescence and production of pro-inflammatory cytokines including IL-1α, IL-1β, IL-6, IL-18 and TNF-α, respectively. Furthermore, Elabela upregulated the protein levels of PPAR-γ in VitD3-overloaded mice. Administrating PPAR-γ inhibitor GW9662 or blocking the efflux of intracellular copper abolished the protective effect of Elabela on vascular calcification by enhancing levels of FDX1, Slc31a1, Runx2, and BMP2. CONCLUSION Elabela plays a crucial role in protecting against vascular cuproptosis and arterial calcification by activating the PPAR-γ /FDX1 signaling. Elabela supplementation and cuproptosis suppression serve as effective therapeutic approaches for managing vascular calcification and related cardiovascular disorders.
Collapse
Affiliation(s)
- Rui-Qiang Qi
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yu-Fei Chen
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing Cheng
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jia-Wei Song
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yi-Hang Chen
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Si-Yuan Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Ying Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Kai-Xin Yan
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Xiao-Yan Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Jing Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Jiu-Chang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China.
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
7
|
Pan X, Song Y, Liang Y, Feng G, Wang Z. Roseburia intestinalis: A possible target for vascular calcification. Heliyon 2024; 10:e39865. [PMID: 39524709 PMCID: PMC11550659 DOI: 10.1016/j.heliyon.2024.e39865] [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: 01/21/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
With the advancement of metagenomics and metabolomics techniques, the crucial role of the gut microbiome in intestinal, cardiovascular, and metabolic disorders has been extensively explored. Vascular calcification (VC) is common in atherosclerosis, hypertension, diabetes mellitus, and chronic kidney disease. Moreover, it is a significant cause of cardiovascular diseases and mortality. Roseburia intestinalis, as a promising candidate for the next generation of probiotics, plays a substantial role in inhibiting the systemic inflammatory response and holds great potential in the treatment of intestinal diseases, cardiovascular diseases, and metabolic disorders. Its primary metabolite, butyrate, acts on specific receptors (GPR43, GPR41, GPR109a). It enters cells via transporters (MCT1, SMCT1), affecting gene expression through HDACs, PPARγ and Nrf2, promoting energy metabolism and changing the concentration of other metabolites (including AGEs, LPS, BHB) in the circulation to affect the body's life activities. In this paper, we focus on the possible mechanism of the primary metabolite butyrate of Roseburia intestinalis in inhibiting VC, which may become a potential therapeutic target for the treatment of VC and the ways to enhance its effect.
Collapse
Affiliation(s)
- Xinyun Pan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| | - Yunjian Song
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| | - Yapeng Liang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Guoquan Feng
- Department of Imaging, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| |
Collapse
|
8
|
Mitsis A, Khattab E, Christodoulou E, Myrianthopoulos K, Myrianthefs M, Tzikas S, Ziakas A, Fragakis N, Kassimis G. From Cells to Plaques: The Molecular Pathways of Coronary Artery Calcification and Disease. J Clin Med 2024; 13:6352. [PMID: 39518492 PMCID: PMC11545949 DOI: 10.3390/jcm13216352] [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: 09/22/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
Coronary artery calcification (CAC) is a hallmark of atherosclerosis and a critical factor in the development and progression of coronary artery disease (CAD). This review aims to address the complex pathophysiological mechanisms underlying CAC and its relationship with CAD. We examine the cellular and molecular processes that drive the formation of calcified plaques, highlighting the roles of inflammation, lipid accumulation, and smooth muscle cell proliferation. Additionally, we explore the genetic and environmental factors that contribute to the heterogeneity in CAC and CAD presentation among individuals. Understanding these intricate mechanisms is essential for developing targeted therapeutic strategies and improving diagnostic accuracy. By integrating current research findings, this review provides a comprehensive overview of the pathways linking CAC to CAD, offering insights into potential interventions to mitigate the burden of these interrelated conditions.
Collapse
Affiliation(s)
- Andreas Mitsis
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Elina Khattab
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Evi Christodoulou
- Cardiology Department, Limassol General Hospital, State Health Services Organization, Limassol 3304, Cyprus;
| | - Kimon Myrianthopoulos
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Michael Myrianthefs
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Stergios Tzikas
- Third Department of Cardiology, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Antonios Ziakas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Nikolaos Fragakis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - George Kassimis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| |
Collapse
|
9
|
Kang JH, Kawano T, Murata M, Toita R. Vascular calcification and cellular signaling pathways as potential therapeutic targets. Life Sci 2024; 336:122309. [PMID: 38042282 DOI: 10.1016/j.lfs.2023.122309] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023]
Abstract
Increased vascular calcification (VC) is observed in patients with cardiovascular diseases such as atherosclerosis, diabetes, and chronic kidney disease. VC is divided into three types according to its location: intimal, medial, and valvular. Various cellular signaling pathways are associated with VC, including the Wnt, mitogen-activated protein kinase, phosphatidylinositol-3 kinase/Akt, cyclic nucleotide-dependent protein kinase, protein kinase C, calcium/calmodulin-dependent kinase II, adenosine monophosphate-activated protein kinase/mammalian target of rapamycin, Ras homologous GTPase, apoptosis, Notch, and cytokine signaling pathways. In this review, we discuss the literature concerning the key cellular signaling pathways associated with VC and their role as potential therapeutic targets. Inhibitors to these pathways represent good candidates for use as potential therapeutic agents for the prevention and treatment of VC.
Collapse
Affiliation(s)
- Jeong-Hun Kang
- National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka 564-8565, Japan.
| | - Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan; AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, AIST, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
10
|
Abstract
Vascular calcification (VC) is recognized as a predictor of all-cause and CVD mortality in chronic kidney disease (CKD). VC in CKD is possibly associated with serum sclerostin. The study systematically investigated the role of serum sclerostin in VC in CKD. Following the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols, a systematic search was performed of the PubMed, Cochrane Library, and EMBASE databases from inception to 11 November 2022, to identify relevant eligible studies. The data were retrieved, analyzed, and summarized. The hazard ratios (HRs) and odds ratios (ORs) with their corresponding confidence intervals (CIs) were derived and pooled. Thirteen reports (3125 patients) met the inclusion criteria and were included. Sclerostin was associated with the presence of VC (pooled OR = 2.75, 95%CI = 1.81-4.19, p < 0.01) and all-cause mortality (pooled HR = 1.22, 95%CI = 1.19-1.25, p < 0.01) among patients with CKD, but with a decreased risk of cardiovascular events (HR = 0.98, 95%CI = 0.97-1.00, p = 0.02). This meta-analysis suggests that serum sclerostin is associated with VC and all-cause mortality among patients with CKD.
Collapse
Affiliation(s)
- Yan Lin
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, China
| | - Liman Mao
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, China
| | - Siqi Chen
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, China
| | - Canxin Zhou
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, China
| |
Collapse
|
11
|
Woo SH, Kim DY, Choi JH. Roles of Vascular Smooth Muscle Cells in Atherosclerotic Calcification. J Lipid Atheroscler 2023; 12:106-118. [PMID: 37265849 PMCID: PMC10232217 DOI: 10.12997/jla.2023.12.2.106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/08/2023] [Accepted: 03/17/2023] [Indexed: 06/03/2023] Open
Abstract
The accumulation of calcium in atherosclerotic plaques is a prominent feature of advanced atherosclerosis, and it has a strong positive correlation with the total burden of atherosclerosis. Atherosclerotic calcification usually appears first at the necrotic core, indicating that cell death and inflammatory processes are involved in calcification. During atherosclerotic inflammation, various cell types, such as vascular smooth muscle cells, nascent resident pericytes, circulating stem cells, or adventitial cells, have been assumed to differentiate into osteoblastic cells, which lead to vascular calcification. Among these cell types, vascular smooth muscle cells are considered a major contributor to osteochondrogenic cells in the atherosclerotic milieu. In this review, we summarize the molecular mechanisms underlying the osteochondrogenic switch of vascular smooth muscle cells in atherosclerotic plaques.
Collapse
Affiliation(s)
- Sang-Ho Woo
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute of Natural Sciences, Research Institute for Convergence of Basic Sciences, Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
| |
Collapse
|
12
|
Akoumianakis I, Polkinghorne M, Antoniades C. Non-canonical WNT signalling in cardiovascular disease: mechanisms and therapeutic implications. Nat Rev Cardiol 2022; 19:783-797. [PMID: 35697779 PMCID: PMC9191761 DOI: 10.1038/s41569-022-00718-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 12/15/2022]
Abstract
WNT signalling comprises a diverse spectrum of receptor-mediated pathways activated by a large family of WNT ligands and influencing fundamental biological processes. WNT signalling includes the β-catenin canonical pathway and the non-canonical pathways, namely the planar cell polarity and the calcium-dependent pathways. Advances over the past decade have linked non-canonical WNT signalling with key mechanisms of atherosclerosis, including oxidative stress, endothelial dysfunction, macrophage activation and vascular smooth muscle cell phenotype regulation. In addition, non-canonical WNT signalling is involved in crucial aspects of myocardial biology, from fibrosis to hypertrophy and oxidative stress. Importantly, non-canonical WNT signalling activation has complex effects in adipose tissue in the context of obesity, thereby potentially linking metabolic and vascular diseases. Tissue-specific targeting of non-canonical WNT signalling might be associated with substantial risks of off-target tumorigenesis, challenging its therapeutic potential. However, novel technologies, such as monoclonal antibodies, recombinant decoy receptors, tissue-specific gene silencing with small interfering RNAs and gene editing with CRISPR-Cas9, might enable more efficient therapeutic targeting of WNT signalling in the cardiovascular system. In this Review, we summarize the components of non-canonical WNT signalling, their links with the main mechanisms of atherosclerosis, heart failure and arrhythmias, and the rationale for targeting individual components of non-canonical WNT signalling for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Murray Polkinghorne
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
13
|
Tripathi M, Singh BK, Liehn EA, Lim SY, Tikno K, Castano-Mayan D, Rattanasopa C, Nilcham P, Abdul Ghani SAB, Wu Z, Azhar SH, Zhou J, Hernández-Resèndiz S, Crespo-Avilan GE, Sinha RA, Farah BL, Moe KT, De Silva DA, Angeli V, Singh MK, Singaraja RR, Hausenloy DJ, Yen PM. Caffeine prevents restenosis and inhibits vascular smooth muscle cell proliferation through the induction of autophagy. Autophagy 2022; 18:2150-2160. [PMID: 35012409 PMCID: PMC9466618 DOI: 10.1080/15548627.2021.2021494] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 01/13/2023] Open
Abstract
Caffeine is among the most highly consumed substances worldwide, and it has been associated with decreased cardiovascular risk. Although caffeine has been shown to inhibit the proliferation of vascular smooth muscle cells (VSMCs), the mechanism underlying this effect is unknown. Here, we demonstrated that caffeine decreased VSMC proliferation and induced macroautophagy/autophagy in an in vivo vascular injury model of restenosis. Furthermore, we studied the effects of caffeine in primary human and mouse aortic VSMCs and immortalized mouse aortic VSMCs. Caffeine decreased cell proliferation, and induced autophagy flux via inhibition of MTOR signaling in these cells. Genetic deletion of the key autophagy gene Atg5, and the Sqstm1/p62 gene encoding a receptor protein, showed that the anti-proliferative effect by caffeine was dependent upon autophagy. Interestingly, caffeine also decreased WNT-signaling and the expression of two WNT target genes, Axin2 and Ccnd1 (cyclin D1). This effect was mediated by autophagic degradation of a key member of the WNT signaling cascade, DVL2, by caffeine to decrease WNT signaling and cell proliferation. SQSTM1/p62, MAP1LC3B-II and DVL2 were also shown to interact with each other, and the overexpression of DVL2 counteracted the inhibition of cell proliferation by caffeine. Taken together, our in vivo and in vitro findings demonstrated that caffeine reduced VSMC proliferation by inhibiting WNT signaling via stimulation of autophagy, thus reducing the vascular restenosis. Our findings suggest that caffeine and other autophagy-inducing drugs may represent novel cardiovascular therapeutic tools to protect against restenosis after angioplasty and/or stent placement.
Collapse
Affiliation(s)
- Madhulika Tripathi
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
| | - Brijesh Kumar Singh
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
| | - Elisa A. Liehn
- National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore-
- Insitute for Molecular Medicine, University of Southern Denmark, Odense, J.B. Winsløws Vej 25, 5230, Odense, Denmark
- Department for Cardiology, Angiology and Intensive Care, Aachen, Germany
| | - Sheau Yng Lim
- Immunology Translational Research Program, Department of Microbiology & Immunology, Immunology Programme, Life Sciences Institute, Singapore- 117456
| | - Keziah Tikno
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
| | - David Castano-Mayan
- Translational Laboratories in Genetic Medicine, A*star Institute, and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chutima Rattanasopa
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
- Translational Laboratories in Genetic Medicine, A*star Institute, and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Pakhwan Nilcham
- Department for Cardiology, Angiology and Intensive Care, Aachen, Germany
| | | | - Zihao Wu
- Translational Laboratories in Genetic Medicine, A*star Institute, and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Syaza Hazwany Azhar
- Immunology Translational Research Program, Department of Microbiology & Immunology, Immunology Programme, Life Sciences Institute, Singapore- 117456
| | - Jin Zhou
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
| | - Sauri Hernández-Resèndiz
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore-
| | - Gustavo E. Crespo-Avilan
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore-
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, India
| | - Benjamin Livingston Farah
- Department of Anatomical Pathology, Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Kyaw Thu Moe
- Newcastle University Medicine Malaysia, Newcastle University, 79200 Gelang Patah, Johor,Malaysia
| | - Deidre Anne De Silva
- Department of Neurology, National Neuroscience Institute, Department of Neurology, Singapore General Hospital, Outram Road, Singapore, 169608
| | - Veronique Angeli
- Immunology Translational Research Program, Department of Microbiology & Immunology, Immunology Programme, Life Sciences Institute, Singapore- 117456
| | - Manvendra K. Singh
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
- National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore-
| | - Roshni R. Singaraja
- Translational Laboratories in Genetic Medicine, A*star Institute, and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Yong Loo Lin School of Medicine, National University, Singapore-117597
| | - Derek J. Hausenloy
- National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore-
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 7 Chenies Mews, Bloomsbury, London WC1E 6HX, United Kingdom
- Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, 500 Liufeng Road, Wufeng District, Taichung City, Taiwan
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Paul Michael Yen
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
- Endocrinology, Diabetes, and Metabolism Division, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
14
|
Awan S, Lambert M, Imtiaz A, Alpy F, Tomasetto C, Oulad-Abdelghani M, Schaeffer C, Moritz C, Julien-David D, Najib S, Martinez LO, Matz RL, Collet X, Silva-Rojas R, Böhm J, Herz J, Terrand J, Boucher P. Wnt5a Promotes Lysosomal Cholesterol Egress and Protects Against Atherosclerosis. Circ Res 2022; 130:184-199. [PMID: 34886684 PMCID: PMC8776607 DOI: 10.1161/circresaha.121.318881] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Impairment of cellular cholesterol trafficking is at the heart of atherosclerotic lesions formation. This involves egress of cholesterol from the lysosomes and 2 lysosomal proteins, the NPC1 (Niemann-Pick C1) and NPC2 that promotes cholesterol trafficking. However, movement of cholesterol out the lysosome and how disrupted cholesterol trafficking leads to atherosclerosis is unclear. As the Wnt ligand, Wnt5a inhibits the intracellular accumulation of cholesterol in multiple cell types, we tested whether Wnt5a interacts with the lysosomal cholesterol export machinery and studied its role in atherosclerotic lesions formation. METHODS We generated mice deleted for the Wnt5a gene in vascular smooth muscle cells. To establish whether Wnt5a also protects against cholesterol accumulation in human vascular smooth muscle cells, we used a CRISPR/Cas9 guided nuclease approach to generate human vascular smooth muscle cells knockout for Wnt5a. RESULTS We show that Wnt5a is a crucial component of the lysosomal cholesterol export machinery. By increasing lysosomal acid lipase expression, decreasing metabolic signaling by the mTORC1 (mechanistic target of rapamycin complex 1) kinase, and through binding to NPC1 and NPC2, Wnt5a senses changes in dietary cholesterol supply and promotes lysosomal cholesterol egress to the endoplasmic reticulum. Consequently, loss of Wnt5a decoupled mTORC1 from variations in lysosomal sterol levels, disrupted lysosomal function, decreased cholesterol content in the endoplasmic reticulum, and promoted atherosclerosis. CONCLUSIONS These results reveal an unexpected function of the Wnt5a pathway as essential for maintaining cholesterol homeostasis in vivo.
Collapse
Affiliation(s)
- Sara Awan
- UMR-S INSERM 1109, University of Strasbourg, 1, place de l’Hôpital, 67000 Strasbourg, France
| | - Magalie Lambert
- UMR-S INSERM 1109, University of Strasbourg, 1, place de l’Hôpital, 67000 Strasbourg, France
| | - Ali Imtiaz
- UMR-S INSERM 1109, University of Strasbourg, 1, place de l’Hôpital, 67000 Strasbourg, France
| | - Fabien Alpy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), University of Strasbourg, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Catherine Tomasetto
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), University of Strasbourg, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Mustapha Oulad-Abdelghani
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), University of Strasbourg, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Christine Schaeffer
- Institut Pluridisciplinaire Hubert Curien, University of Strasbourg, 23 Rue du Loess, 67037 Strasbourg, France
| | - Chloé Moritz
- Institut Pluridisciplinaire Hubert Curien, University of Strasbourg, 23 Rue du Loess, 67037 Strasbourg, France
| | - Diane Julien-David
- CNRS, UMR 7178, University of Strasbourg, 23 Rue du Loess, 67037 Strasbourg, France
| | - Souad Najib
- Institute of Metabolic and Cardiovascular Diseases, I2MC, INSERM, UMR, 1048, 1 avenue du Professeur Jean Poulhès, 31432 Toulouse, France
| | - Laurent O. Martinez
- Institute of Metabolic and Cardiovascular Diseases, I2MC, INSERM, UMR, 1048, 1 avenue du Professeur Jean Poulhès, 31432 Toulouse, France
| | - Rachel L. Matz
- UMR-S INSERM 1109, University of Strasbourg, 1, place de l’Hôpital, 67000 Strasbourg, France
| | - Xavier Collet
- Institute of Metabolic and Cardiovascular Diseases, I2MC, INSERM, UMR, 1048, 1 avenue du Professeur Jean Poulhès, 31432 Toulouse, France
| | - Roberto Silva-Rojas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), University of Strasbourg, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Johann Böhm
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), University of Strasbourg, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
| | - Jérôme Terrand
- UMR-S INSERM 1109, University of Strasbourg, 1, place de l’Hôpital, 67000 Strasbourg, France
| | - Philippe Boucher
- UMR-S INSERM 1109, University of Strasbourg, 1, place de l’Hôpital, 67000 Strasbourg, France
| |
Collapse
|
15
|
Luo M, Sun W, Kong X. Emodin alleviates aortic valvular calcification by inhibiting the AKT/FOXO1 pathway. Ann Anat 2021; 240:151885. [PMID: 34958913 DOI: 10.1016/j.aanat.2021.151885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Valvular calcification commonly occurs in elderly individuals, and is increasingly considered an important economic and health burden. However, no efficient drugs against valvular calcification are available. The present work aimed to examine emodin's suppressive effect on high-calcium-dependent valve calcification and explore the underpinning mechanisms. METHODS Experiments were carried out in mice receiving vitamin D (Vit D) to induce valvular calcification. RESULTS Cell viability and apoptosis assays demonstrated celastrol suppressed proliferation and increased apoptosis in porcine aortic valve interstitial cells (PAVICs) at concentrations higher than 10 μM. Emodin (5 μM) attenuated the upregulation of osteogenic genes as well as calcium accumulation in PAVICs under high-calcium conditions. The elevations of calcium content in serum and valve, and calcium accumulation in valve and artery were suppressed by emodin in mice with valvular calcification after joint treatment with adenine and Vit D. In addition, p-AKT and p-FOXO1 were upregulated in PAVICs under high-calcium conditions, and this effect was reversed by emodin treatment. SC79, an AKT activator, reversed emodin's suppressive effects on increased calcium content, calcium deposition and osteogenic gene expression in PAVICs induced by calcific medium. CONCLUSIONS These data demonstrated emodin alleviates high-calcium-associated valvular calcification via AKT/FOXO1 signaling suppression, providing new insights into therapeutic strategies for clinical valvular calcification.
Collapse
Affiliation(s)
- Man Luo
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Sun
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiangqing Kong
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
16
|
Cannata-Andía JB, Carrillo-López N, Messina OD, Hamdy NAT, Panizo S, Ferrari SL, on behalf of the International Osteoporosis Foundation (IOF) Working Group on Bone and Cardiovascular Diseases. Pathophysiology of Vascular Calcification and Bone Loss: Linked Disorders of Ageing? Nutrients 2021; 13:3835. [PMID: 34836090 PMCID: PMC8623966 DOI: 10.3390/nu13113835] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
Vascular Calcification (VC), low bone mass and fragility fractures are frequently observed in ageing subjects. Although this clinical observation could be the mere coincidence of frequent age-dependent disorders, clinical and experimental data suggest that VC and bone loss could share pathophysiological mechanisms. Indeed, VC is an active process of calcium and phosphate precipitation that involves the transition of the vascular smooth muscle cells (VSMCs) into osteoblast-like cells. Among the molecules involved in this process, parathyroid hormone (PTH) plays a key role acting through several mechanisms which includes the regulation of the RANK/RANKL/OPG system and the Wnt/ß-catenin pathway, the main pathways for bone resorption and bone formation, respectively. Furthermore, some microRNAs have been implicated as common regulators of bone metabolism, VC, left ventricle hypertrophy and myocardial fibrosis. Elucidating the common mechanisms between ageing; VC and bone loss could help to better understand the potential effects of osteoporosis drugs on the CV system.
Collapse
Affiliation(s)
- Jorge B. Cannata-Andía
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (N.C.-L.); (S.P.)
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (N.C.-L.); (S.P.)
| | - Osvaldo D. Messina
- Investigaciones Reumatológicas y Osteológicas (IRO), Buenos Aires 1114, Argentina;
| | - Neveen A. T. Hamdy
- Center for Bone Quality, Division Endocrinology, Department of Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Sara Panizo
- Bone and Mineral Research Unit, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Retic REDinREN-ISCIII, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (N.C.-L.); (S.P.)
| | - Serge L. Ferrari
- Service and Laboratory of Bone Diseases, Department of Medicine, Faculty of Medicine, Geneva University Hospital, 1211 Geneva, Switzerland;
| | | |
Collapse
|
17
|
Chinetti G, Neels JG. Roles of Nuclear Receptors in Vascular Calcification. Int J Mol Sci 2021; 22:6491. [PMID: 34204304 PMCID: PMC8235358 DOI: 10.3390/ijms22126491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
Vascular calcification is defined as an inappropriate accumulation of calcium depots occurring in soft tissues, including the vascular wall. Growing evidence suggests that vascular calcification is an actively regulated process, sharing similar mechanisms with bone formation, implicating both inhibitory and inducible factors, mediated by osteoclast-like and osteoblast-like cells, respectively. This process, which occurs in nearly all the arterial beds and in both the medial and intimal layers, mainly involves vascular smooth muscle cells. In the vascular wall, calcification can have different clinical consequences, depending on the pattern, localization and nature of calcium deposition. Nuclear receptors are transcription factors widely expressed, activated by specific ligands that control the expression of target genes involved in a multitude of pathophysiological processes, including metabolism, cancer, inflammation and cell differentiation. Some of them act as drug targets. In this review we describe and discuss the role of different nuclear receptors in the control of vascular calcification.
Collapse
Affiliation(s)
- Giulia Chinetti
- Université Côte d’Azur, CHU, INSERM, C3M, 06204 Nice, France;
| | - Jaap G. Neels
- Université Côte d’Azur, INSERM, C3M, 06204 Nice, France
| |
Collapse
|
18
|
Carrillo-López N, Martínez-Arias L, Fernández-Villabrille S, Ruiz-Torres MP, Dusso A, Cannata-Andía JB, Naves-Díaz M, Panizo S. Role of the RANK/RANKL/OPG and Wnt/β-Catenin Systems in CKD Bone and Cardiovascular Disorders. Calcif Tissue Int 2021; 108:439-451. [PMID: 33586001 DOI: 10.1007/s00223-020-00803-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/19/2020] [Indexed: 12/23/2022]
Abstract
In the course of chronic kidney disease (CKD), alterations in the bone-vascular axis augment the risk of bone loss, fractures, vascular and soft tissue calcification, left ventricular hypertrophy, renal and myocardial fibrosis, which markedly increase morbidity and mortality rates. A major challenge to improve skeletal and cardiovascular outcomes in CKD patients requires a better understanding of the increasing complex interactions among the main modulators of the bone-vascular axis. Serum parathyroid hormone (PTH), phosphorus (P), calcium (Ca), fibroblast growth factor 23 (FGF23), calcidiol, calcitriol and Klotho are involved in this axis interact with RANK/RANKL/OPG system and the Wnt/β-catenin pathway. The RANK/RANKL/OPG system controls bone remodeling by inducing osteoblast synthesis of RANKL and downregulating OPG production and it is also implicated in vascular calcification. The complexity of this system has recently increased due the discovery of LGR4, a novel RANKL receptor involved in bone formation, but possibly also in vascular calcification. The Wnt/β-catenin pathway plays a key role in bone formation: when this pathway is activated, bone is formed, but when it is inhibited, bone formation is stopped. In the progression of CKD, a downregulation of the Wnt/β-catenin pathway has been described which occurs mainly through the not coincident elevations of sclerostin, Dickkopf1 (Dkk1) and the secreted Frizzled Related Proteins (sFRPs). This review analyzes the interactions of PTH, P, Ca, FGF23, calcidiol, calcitriol and Klotho with the RANKL/RANKL/OPG system and the Wnt/β-catenin, pathway and their implications in bone and cardiovascular disorders in CKD.
Collapse
Affiliation(s)
- Natalia Carrillo-López
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain
| | - Laura Martínez-Arias
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain
| | - Sara Fernández-Villabrille
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain
| | - María Piedad Ruiz-Torres
- Department of System Biology, Universidad de Alcalá, Retic REDinREN-ISCIII, Alcalá de Henares, Spain
| | - Adriana Dusso
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain
| | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain.
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain
| | - Sara Panizo
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain.
| | | |
Collapse
|
19
|
Tang N, Li H, Zhang L, Zhang X, Chen Y, Shou H, Feng S, Chen X, Luo Y, Tang R, Wang B. A Macromolecular Drug for Cancer Therapy via Extracellular Calcification. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202016122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Ning Tang
- Cancer Institute The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 China
- Institute of Translational Medicine Zhejiang University Hangzhou 310029 China
| | - Hanhui Li
- Cancer Institute The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 China
- Institute of Translational Medicine Zhejiang University Hangzhou 310029 China
| | - Lihong Zhang
- Cancer Institute The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 China
- Department of Biochemistry Zhejiang University School of Medicine Hangzhou 310058 China
| | - Xueyun Zhang
- Cancer Institute The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 China
- Department of Biochemistry Zhejiang University School of Medicine Hangzhou 310058 China
| | - Yanni Chen
- Cancer Institute The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 China
- Institute of Translational Medicine Zhejiang University Hangzhou 310029 China
| | - Hao Shou
- Cancer Institute The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 China
- Institute of Translational Medicine Zhejiang University Hangzhou 310029 China
| | - Shuaishuai Feng
- Cancer Institute The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 China
- Institute of Translational Medicine Zhejiang University Hangzhou 310029 China
| | - Xinhua Chen
- Department of Hepatobiliary and Pancreatic Surgery Key Laboratory of Combined Multi-organ Transplantation Ministry of Public Health The First Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310003 China
| | - Yan Luo
- Cancer Institute The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 China
- Department of Biochemistry Zhejiang University School of Medicine Hangzhou 310058 China
| | - Ruikang Tang
- Department of Chemistry Zhejiang University Hangzhou 310027 China
| | - Ben Wang
- Cancer Institute The Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou 310009 China
- Institute of Translational Medicine Zhejiang University Hangzhou 310029 China
| |
Collapse
|
20
|
Han Y, Zhang J, Huang S, Cheng N, Zhang C, Li Y, Wang X, Liu J, You B, Du J. MicroRNA-223-3p inhibits vascular calcification and the osteogenic switch of vascular smooth muscle cells. J Biol Chem 2021; 296:100483. [PMID: 33647318 PMCID: PMC8039724 DOI: 10.1016/j.jbc.2021.100483] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/19/2021] [Accepted: 02/25/2021] [Indexed: 11/20/2022] Open
Abstract
Vascular calcification is the ectopic deposition of calcium hydroxyapatite minerals in arterial wall, which involves the transdifferentiation of vascular smooth muscle cells (VSMCs) toward an osteogenic phenotype. However, the underlying molecular mechanisms regulating the VSMC osteogenic switch remain incompletely understood. In this study, we examined the roles of microRNAs (miRNAs) in vascular calcification. miRNA-seq transcriptome analysis identified miR-223-3p as a candidate miRNA in calcified mouse aortas. MiR-223-3p knockout aggravated calcification in both medial and atherosclerotic vascular calcification models. Further, RNA-seq transcriptome analysis verified JAK-STAT and PPAR signaling pathways were upregulated in both medial and atherosclerotic calcified aortas. Overlapping genes in these signaling pathways with predicted target genes of miR-223-3p derived from miRNA databases, we identified signal transducer and activator of transcription 3 (STAT3) as a potential target gene of miR-223-3p in vascular calcification. In vitro experiments showed that miR-223-3p blocked interleukin-6 (IL-6)/STAT3 signaling, thereby preventing the osteogenic switch and calcification of VSMCs. In contrast, overexpression of STAT3 diminished the effect of miR-223-3p. Taken together, the results indicate a protective role of miR-223-3p that inhibits both medial and atherosclerotic vascular calcification by regulating IL-6/STAT3 signaling-mediated VSMC transdifferentiation.
Collapse
Affiliation(s)
- Yingchun Han
- Beijing Anzhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Jichao Zhang
- Beijing Anzhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Shan Huang
- Beijing Anzhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China; School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Naixuan Cheng
- Beijing Anzhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China; School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Congcong Zhang
- Beijing Anzhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Yulin Li
- Beijing Anzhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Xiaonan Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Jinghua Liu
- Beijing Anzhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Bin You
- Beijing Anzhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Jie Du
- Beijing Anzhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China.
| |
Collapse
|
21
|
Tang N, Li H, Zhang L, Zhang X, Chen Y, Shou H, Feng S, Chen X, Luo Y, Tang R, Wang B. A Macromolecular Drug for Cancer Therapy via Extracellular Calcification. Angew Chem Int Ed Engl 2021; 60:6509-6517. [PMID: 33427367 DOI: 10.1002/anie.202016122] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Indexed: 12/15/2022]
Abstract
Cancer chemotherapy typically relies on drug endocytosis and inhibits tumor cell proliferation via intracellular pathways; however, severe side effects may arise. In this study, we performed a first attempt to develop macromolecular-induced extracellular chemotherapy involving biomineralization by absorbing calcium from the blood through a new type of drug, polysialic acid conjugated with folate (folate-polySia), which selectively induces biogenic mineral formation on tumor cells and results in the pathological calcification of tumors. The macromolecule-initiated extracellular calcification causes cancer cell death mainly by intervening with the glycolysis process in cancer cells. Systemic administration of folate-polySia inhibited cervical and breast tumor growth and dramatically improved survival rates in mice. This study provides an extracellular therapeutic approach for malignant tumor diseases via calcification that is ready for clinical trials and offers new insights into macromolecular anticancer drug discovery.
Collapse
Affiliation(s)
- Ning Tang
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Hanhui Li
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Lihong Zhang
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xueyun Zhang
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yanni Chen
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Hao Shou
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Shuaishuai Feng
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Xinhua Chen
- Department of Hepatobiliary and Pancreatic Surgery, Key Laboratory of Combined Multi-organ Transplantation Ministry of Public Health, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yan Luo
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, 310027, China
| | - Ben Wang
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| |
Collapse
|
22
|
Wang L, Chennupati R, Jin YJ, Li R, Wang S, Günther S, Offermanns S. YAP/TAZ Are Required to Suppress Osteogenic Differentiation of Vascular Smooth Muscle Cells. iScience 2020; 23:101860. [PMID: 33319178 PMCID: PMC7726335 DOI: 10.1016/j.isci.2020.101860] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/10/2020] [Accepted: 11/20/2020] [Indexed: 12/22/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) represent the prevailing cell type of arterial vessels and are essential for blood vessel structure and homeostasis. They have substantial potential for phenotypic plasticity when exposed to various stimuli in their local microenvironment. How VSMCs maintain their differentiated contractile phenotype is still poorly understood. Here we demonstrate that the Hippo pathway effectors YAP and TAZ play a critical role in maintaining the differentiated contractile phenotype of VSMCs. In the absence of YAP/TAZ, VSMCs lose their differentiated phenotype and undergo osteogenic differentiation, which results in vascular calcification. Osteogenic transdifferentiation was accompanied by the upregulation of Wnt target genes. The absence of YAP/TAZ in VSMCs led to Disheveled 3 (DVL3) nuclear translocation and upregulation of osteogenesis-associated genes independent of canonical Wnt/β-catenin signaling activation. Our data indicate that cytoplasmic YAP/TAZ interact with DVL3 to avoid its nuclear translocation and osteogenic differentiation, thereby maintaining the differentiated phenotype of VSMCs. YAP/TAZ play an important role in maintaining vascular SMCs contractile phenotype Loss of YAP/TAZ in vSMCs leads to reduced expression of smooth muscle marker genes Loss of YAP/TAZ in vSMCs results in reduced artery contractility Deficiency of YAP/TAZ in vSMCs leads to osteogenic transdifferentiation
Collapse
Affiliation(s)
- Lei Wang
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Ramesh Chennupati
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Young-June Jin
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - Rui Li
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany
| | - ShengPeng Wang
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany.,Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Yanta District, Xi'an 710061, China
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim 61231, Germany.,Center for Molecular Medicine, Medical Faculty, Goethe University, Frankfurt am Main 60590, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Frankfurt Rhine-Main, 13347 Berlin, Germany
| |
Collapse
|
23
|
Reinhold S, Blankesteijn WM, Foulquier S. The Interplay of WNT and PPARγ Signaling in Vascular Calcification. Cells 2020; 9:cells9122658. [PMID: 33322009 PMCID: PMC7763279 DOI: 10.3390/cells9122658] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/02/2022] Open
Abstract
Vascular calcification (VC), the ectopic deposition of calcium phosphate crystals in the vessel wall, is one of the primary contributors to cardiovascular death. The pathology of VC is determined by vascular topography, pre-existing diseases, and our genetic heritage. VC evolves from inflammation, mediated by macrophages, and from the osteochondrogenic transition of vascular smooth muscle cells (VSMC) in the atherosclerotic plaque. This pathologic transition partly resembles endochondral ossification, involving the chronologically ordered activation of the β-catenin-independent and -dependent Wingless and Int-1 (WNT) pathways and the termination of peroxisome proliferator-activated receptor γ (PPARγ) signal transduction. Several atherosclerotic plaque studies confirmed the differential activity of PPARγ and the WNT signaling pathways in VC. Notably, the actively regulated β-catenin-dependent and -independent WNT signals increase the osteochondrogenic transformation of VSMC through the up-regulation of the osteochondrogenic transcription factors SRY-box transcription factor 9 (SOX9) and runt-related transcription factor 2 (RUNX2). In addition, we have reported studies showing that WNT signaling pathways may be antagonized by PPARγ activation via the expression of different families of WNT inhibitors and through its direct interaction with β-catenin. In this review, we summarize the existing knowledge on WNT and PPARγ signaling and their interplay during the osteochondrogenic differentiation of VSMC in VC. Finally, we discuss knowledge gaps on this interplay and its possible clinical impact.
Collapse
Affiliation(s)
- Stefan Reinhold
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (S.R.); (W.M.B.)
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (S.R.); (W.M.B.)
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands; (S.R.); (W.M.B.)
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-433881409
| |
Collapse
|
24
|
Wang W, Liu X, Zheng X, Jin HJ, Li X. Biomineralization: An Opportunity and Challenge of Nanoparticle Drug Delivery Systems for Cancer Therapy. Adv Healthc Mater 2020; 9:e2001117. [PMID: 33043640 DOI: 10.1002/adhm.202001117] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/29/2020] [Indexed: 12/12/2022]
Abstract
Biomineralization is a common process in organisms to produce hard biomaterials by combining inorganic ions with biomacromolecules. Multifunctional nanoplatforms are developed based on the mechanism of biomineralization in many biomedical applications. In the past few years, biomineralization-based nanoparticle drug delivery systems for the cancer treatment have gained a lot of research attention due to the advantages including simple preparation, good biocompatibility, degradability, easy modification, versatility, and targeting. In this review, the research trends of biomineralization-based nanoparticle drug delivery systems and their applications in cancer therapy are summarized. This work aims to promote future researches on cancer therapy based on biomineralization. Rational design of nanoparticle drug delivery systems can overcome the bottleneck in the clinical transformation of nanomaterials. At the same time, biomineralization has also provided new research ideas for cancer treatment, i.e., targeted therapy, which has significantly better performance.
Collapse
Affiliation(s)
- Weicai Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Xiaofan Liu
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Xiangjiang Zheng
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Hyung Jong Jin
- Department of Bioscience and Biotechnology The University of Suwon Hwaseong Gyeonggi‐Do 18323 Republic of Korea
| | - Xuemei Li
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| |
Collapse
|
25
|
Extracellular Nucleotides Regulate Arterial Calcification by Activating Both Independent and Dependent Purinergic Receptor Signaling Pathways. Int J Mol Sci 2020; 21:ijms21207636. [PMID: 33076470 PMCID: PMC7589647 DOI: 10.3390/ijms21207636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 01/02/2023] Open
Abstract
Arterial calcification, the deposition of calcium-phosphate crystals in the extracellular matrix, resembles physiological bone mineralization. It is well-known that extracellular nucleotides regulate bone homeostasis raising an emerging interest in the role of these molecules on arterial calcification. The purinergic independent pathway involves the enzymes ecto-nucleotide pyrophosphatase/phosphodiesterases (NPPs), ecto-nucleoside triphosphate diphosphohydrolases (NTPDases), 5′-nucleotidase and alkaline phosphatase. These regulate the production and breakdown of the calcification inhibitor—pyrophosphate and the calcification stimulator—inorganic phosphate, from extracellular nucleotides. Maintaining ecto-nucleotidase activities in a well-defined range is indispensable as enzymatic hyper- and hypo-expression has been linked to arterial calcification. The purinergic signaling dependent pathway focusses on the activation of purinergic receptors (P1, P2X and P2Y) by extracellular nucleotides. These receptors influence arterial calcification by interfering with the key molecular mechanisms underlying this pathology, including the osteogenic switch and apoptosis of vascular cells and possibly, by favoring the phenotypic switch of vascular cells towards an adipogenic phenotype, a recent, novel hypothesis explaining the systemic prevention of arterial calcification. Selective compounds influencing the activity of ecto-nucleotidases and purinergic receptors, have recently been developed to treat arterial calcification. However, adverse side-effects on bone mineralization are possible as these compounds reasonably could interfere with physiological bone mineralization.
Collapse
|
26
|
Khan K, Yu B, Kiwan C, Shalal Y, Filimon S, Cipro M, Shum-Tim D, Cecere R, Schwertani A. The Role of Wnt/β-Catenin Pathway Mediators in Aortic Valve Stenosis. Front Cell Dev Biol 2020; 8:862. [PMID: 33015048 PMCID: PMC7513845 DOI: 10.3389/fcell.2020.00862] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023] Open
Abstract
Aortic valve stenosis (AVS) is a prevailing and life-threatening cardiovascular disease in adults over 75 years of age. However, the molecular mechanisms governing the pathogenesis of AVS are yet to be fully unraveled. With accumulating evidence that Wnt signaling plays a key role in the development of AVS, the involvement of Wnt molecules has become an integral study target in AVS pathogenesis. Thus, we hypothesized that the Wnt/β-catenin pathway mediators, SFRP2, DVL2, GSK3β and β-catenin are dysregulated in patients with AVS. Using immunohistochemistry, Real-Time qPCR and Western blotting, we investigated the presence of SFRP2, GSK-3β, DVL2, and β-catenin in normal and stenotic human aortic valves. Markedly higher mRNA and protein expression of GSK-3β, DVL2, β-catenin and SFRP2 were found in stenotic aortic valves. This was further corroborated by observation of their abundant immunostaining, which displayed strong immunoreactivity in diseased aortic valves. Proteomic analyses of selective GSK3b inhibition in calcifying human aortic valve interstitial cells (HAVICs) revealed enrichment of proteins involved organophosphate metabolism, while reducing the activation of pathogenic biomolecular processes. Lastly, use of the potent calcification inhibitor, Fetuin A, in calcifying HAVICs significantly reduced the expression of Wnt signaling genes Wnt3a, Wnt5a, Wnt5b, and Wnt11. The current findings of altered expression of canonical Wnt signaling in AVS suggest a possible role for regulatory Wnts in AVS. Hence, future studies focused on targeting these molecules are warranted to underline their role in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Kashif Khan
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Bin Yu
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Chrystina Kiwan
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Yousif Shalal
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Sabin Filimon
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Megan Cipro
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Dominique Shum-Tim
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Renzo Cecere
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, QC, Canada
| | - Adel Schwertani
- Division of Cardiology and Cardiac Surgery, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
27
|
Kim JA, Choi KM. Newly Discovered Adipokines: Pathophysiological Link Between Obesity and Cardiometabolic Disorders. Front Physiol 2020; 11:568800. [PMID: 32982804 PMCID: PMC7492654 DOI: 10.3389/fphys.2020.568800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/14/2020] [Indexed: 12/21/2022] Open
Abstract
With the increasing prevalence of obesity, obesity-related problems such as cardiometabolic disorders (CMD), are also rapidly increasing. To prevent and alleviate the progressive course of CMD, it is important to discover the pathophysiological mechanisms between obesity and CMD. Adipose tissue is now recognized as an active endocrine organ that releases adipokines. Adipokines play a pivotal role in chronic low-grade inflammation, oxidative stress, and impaired insulin signaling, contributing to metabolic derangement and leading to CMD. Recent studies have provided substantial evidence supporting the association between adipokines and CMD. In this review, we highlight the pathophysiological action of adipokines in CMD that includes metabolic syndrome, type 2 diabetes, non-alcoholic fatty liver disease, and cardiovascular diseases. We focused on translational and clinical research of novel adipokines associated with metabolic and cardiovascular regulation. Exploration of the role of these adipokines connecting obesity and CMD may provide a perspective on adipokine-based therapeutic implications for CMD.
Collapse
Affiliation(s)
- Jung A Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, South Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
28
|
Saito Y, Nakamura K, Ito H. Effects of Eicosapentaenoic Acid on Arterial Calcification. Int J Mol Sci 2020; 21:ijms21155455. [PMID: 32751754 PMCID: PMC7432365 DOI: 10.3390/ijms21155455] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 12/26/2022] Open
Abstract
Arterial calcification is a hallmark of advanced atherosclerosis and predicts cardiovascular events. However, there is no clinically accepted therapy that prevents progression of arterial calcification. HMG-CoA reductase inhibitors, statins, lower low-density lipoprotein-cholesterol and reduce cardiovascular events, but coronary artery calcification is actually promoted by statins. The addition of eicosapentaenoic acid (EPA) to statins further reduced cardiovascular events in clinical trials, JELIS and REDUCE-IT. Additionally, we found that EPA significantly suppressed arterial calcification in vitro and in vivo via suppression of inflammatory responses, oxidative stress and Wnt signaling. However, so far there is a lack of evidence showing the effect of EPA on arterial calcification in a clinical situation. We reviewed the molecular mechanisms of the inhibitory effect of EPA on arterial calcification and the results of some clinical trials.
Collapse
|
29
|
De Maré A, D’Haese PC, Verhulst A. The Role of Sclerostin in Bone and Ectopic Calcification. Int J Mol Sci 2020; 21:ijms21093199. [PMID: 32366042 PMCID: PMC7246472 DOI: 10.3390/ijms21093199] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023] Open
Abstract
Sclerostin, a 22-kDa glycoprotein that is mainly secreted by the osteocytes, is a soluble inhibitor of canonical Wnt signaling. Therefore, when present at increased concentrations, it leads to an increased bone resorption and decreased bone formation. Serum sclerostin levels are known to be increased in the elderly and in patients with chronic kidney disease. In these patient populations, there is a high incidence of ectopic cardiovascular calcification. These calcifications are strongly associated with cardiovascular morbidity and mortality. Although data are still controversial, it is likely that there is a link between ectopic calcification and serum sclerostin levels. The main question, however, remains whether sclerostin exerts either a protective or deleterious role in the ectopic calcification process.
Collapse
|
30
|
Boucher P, Matz RL, Terrand J. atherosclerosis: gone with the Wnt? Atherosclerosis 2020; 301:15-22. [PMID: 32289618 DOI: 10.1016/j.atherosclerosis.2020.03.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/19/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022]
Abstract
Atherosclerosis, a pathology affecting large and medium-sized arteries, is the major cause of cardiovascular morbidity/mortality in industrialized countries. During atherosclerosis, cells accumulate large amounts of cholesterol through the uptake of modified low-density lipoprotein particles to form foam cells. This accumulation forms the basis for the development of the disease and for a large spectrum of other diseases in various organs. Massive research efforts have yielded valuable information about the underlying molecular mechanisms of atherosclerosis. In particular, newer discoveries on the early stage of lesion formation, cholesterol accumulation, reverse cholesterol transport, and local inflammation in the vascular wall have opened unanticipated horizons of understanding and raised novel questions and therapeutic opportunities. In this review, we focus on Wnt signaling, which has received little attention so far, yet affects lysosomal function and signalling pathways that limit cholesterol accumulation. This occurs in different tissues and cell types, including smooth muscle cells, endothelial cells and macrophages in the arterial wall, and thus profoundly impacts on atherosclerotic disease development and progression.
Collapse
Affiliation(s)
- Philippe Boucher
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France.
| | - Rachel L Matz
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France
| | - Jérôme Terrand
- CNRS, UMR 7021, University of Strasbourg, 67401, Illkirch, France
| |
Collapse
|
31
|
Wang D, Zhang Y, Shen C. Research update on the association between SFRP5, an anti-inflammatory adipokine, with obesity, type 2 diabetes mellitus and coronary heart disease. J Cell Mol Med 2020; 24:2730-2735. [PMID: 32004418 PMCID: PMC7077606 DOI: 10.1111/jcmm.15023] [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: 10/18/2019] [Revised: 12/18/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Secreted frizzled-related protein 5 (SFRP5), an anti-inflammatory adipokine secreted by adipocytes, has been demonstrated to exert its anti-inflammatory effect via antagonizing the non-canonical wingless-type family member 5A (WNT5A) signalling pathways. The WNT5A protein, as a potent pro-inflammatory signalling molecule, is strongly involved in a variety of inflammatory disorders such as obesity, type 2 diabetes mellitus (T2DM) and atherosclerosis. In this review, we systematically outlined the current understanding on the roles of SFRP5 in the pathogenesis of three inflammatory diseases including obesity, T2DM and coronary heart disease (CHD). Our review might stimulate future research using SFRP5 as a promising novel therapeutic target for the treatment of obesity, T2DM and CHD.
Collapse
Affiliation(s)
- Di Wang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yaping Zhang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW This review addresses recent developments in studies of lipid regulation of calcific disease of arteries and cardiac valves, including the role of nuclear receptors. The role of lipid-soluble signals and their receptors is timely given the recent evidence and concerns that lipid-lowering treatment may increase the rate of progression of coronary artery calcification, which has been long associated with increased cardiovascular risk. Understanding the mechanisms will be important for interpreting such clinical information. RECENT FINDINGS New findings support regulation of calcific vascular and valvular disease by nuclear receptors, including the vitamin D receptor, glucocorticoid receptor, nutrient-sensing nuclear receptors (liver X receptor, farnesoid X receptor, and peroxisome proliferator-activated receptors), and sex hormone (estrogen and androgen) receptors. There were two major unexpected findings: first, vitamin D supplementation, which was previously believed to prevent or reduce vascular calcification, showed no cardiovascular benefit in large randomized, controlled trials. Second, both epidemiological studies and coronary intravascular ultrasound studies suggest that treatment with HMG-CoA reductase inhibitors increases progression of coronary artery calcification, raising a question of whether there are mechanically stable and unstable forms of coronary calcification. SUMMARY For clinical practice and research, these new findings offer new fundamental mechanisms for vascular calcification and provide new cautionary insights for therapeutic avenues.
Collapse
Affiliation(s)
- Tamer Sallam
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095-1679
| | - Yin Tintut
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Department of Orthopaedic Surgery, University of California, Los Angeles, Los Angeles, CA 90095-1679
| | - Linda L. Demer
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Department of Physiology, University of California, Los Angeles, Los Angeles, CA 90095-1679
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095-1679
| |
Collapse
|
33
|
van der Laan SW, Siemelink MA, Haitjema S, Foroughi Asl H, Perisic L, Mokry M, van Setten J, Malik R, Dichgans M, Worrall BB, Samani NJ, Schunkert H, Erdmann J, Hedin U, Paulsson-Berne G, Björkegrenn JLM, de Borst GJ, Asselbergs FW, den Ruijter FW, de Bakker PIW, Pasterkamp G. Genetic Susceptibility Loci for Cardiovascular Disease and Their Impact on Atherosclerotic Plaques. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002115. [PMID: 30354329 PMCID: PMC7664607 DOI: 10.1161/circgen.118.002115] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Atherosclerosis is a chronic inflammatory disease in part caused by lipid uptake in the vascular wall, but the exact underlying mechanisms leading to acute myocardial infarction and stroke remain poorly understood. Large consortia identified genetic susceptibility loci that associate with large artery ischemic stroke and coronary artery disease. However, deciphering their underlying mechanisms are challenging. Histological studies identified destabilizing characteristics in human atherosclerotic plaques that associate with clinical outcome. To what extent established susceptibility loci for large artery ischemic stroke and coronary artery disease relate to plaque characteristics is thus far unknown but may point to novel mechanisms. Methods: We studied the associations of 61 established cardiovascular risk loci with 7 histological plaque characteristics assessed in 1443 carotid plaque specimens from the Athero-Express Biobank Study. We also assessed if the genotyped cardiovascular risk loci impact the tissue-specific gene expression in 2 independent biobanks, Biobank of Karolinska Endarterectomy and Stockholm Atherosclerosis Gene Expression. Results: A total of 21 established risk variants (out of 61) nominally associated to a plaque characteristic. One variant (rs12539895, risk allele A) at 7q22 associated to a reduction of intraplaque fat, P=5.09×10−6 after correction for multiple testing. We further characterized this 7q22 Locus and show tissue-specific effects of rs12539895 on HBP1 expression in plaques and COG5 expression in whole blood and provide data from public resources showing an association with decreased LDL (low-density lipoprotein) and increase HDL (high-density lipoprotein) in the blood. Conclusions: Our study supports the view that cardiovascular susceptibility loci may exert their effect by influencing the atherosclerotic plaque characteristics.
Collapse
Affiliation(s)
- Sander W van der Laan
- Laboratory of Experimental Cardiology, Division Heart and Lungs, University Medical Center Utrecht, University Utrecht, The Netherlands (S.W.v.d.L., M.A.S., S.H., H.M.d.R., G.P.)
| | - Marten A Siemelink
- Laboratory of Experimental Cardiology, Division Heart and Lungs, University Medical Center Utrecht, University Utrecht, The Netherlands (S.W.v.d.L., M.A.S., S.H., H.M.d.R., G.P.).,Department of Clinical Genetics, University Medical Center Utrecht, University Utrecht, The Netherlands (M.A.S.)
| | - Saskia Haitjema
- Laboratory of Experimental Cardiology, Division Heart and Lungs, University Medical Center Utrecht, University Utrecht, The Netherlands (S.W.v.d.L., M.A.S., S.H., H.M.d.R., G.P.)
| | - Hassan Foroughi Asl
- Cardiovascular Genomics Group, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.F.A.)
| | - Ljubica Perisic
- Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden (L.P., U.H.)
| | - Michal Mokry
- Department of Pediatrics, Wilhelmina Children's Hospital, University Medical Center Utrecht, University Utrecht, The Netherlands (M.M.).,Regenerative Medicine Center Utrecht, University Medical Center Utrecht, University Utrecht, The Netherlands (M.M.)
| | - Jessica van Setten
- Department of Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, University Utrecht, The Netherlands (F.W.A., J.v.S.)
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany (R.M., M.D.)
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany (R.M., M.D.).,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M.D.)
| | - Bradford B Worrall
- Departments of Neurology and Public Health Sciences, University of Virginia, Charlottesville (B.B.W.)
| | | | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester (N.J.S.).,NIHR Leicester Biomedical Research Unit Centre, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, United Kingdom (N.J.S.)
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik an der TU München, Munich Heart Alliance (DZHK), Germany (H.S., J.E.)
| | - Jeanette Erdmann
- Deutsches Herzzentrum München, Klinik an der TU München, Munich Heart Alliance (DZHK), Germany (H.S., J.E.)
| | - Ulf Hedin
- Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden (L.P., U.H.)
| | - Gabrielle Paulsson-Berne
- Unit of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden (G.P.-B.)
| | - Johan L M Björkegrenn
- CMM, Karolinska Institutet, Stockholm, Sweden. Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York (J.L.M.B.).,Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden (J.L.M.B.).,Clinical Gene Networks AB, Stockholm,Sweden (J.L.M.B.)
| | - Gert J de Borst
- Division of Surgical Specialties, Department of Surgery, University Medical Center Utrecht, University Utrecht, The Netherlands (G.J.d.B.)
| | - Folkert W Asselbergs
- Department of Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, University Utrecht, The Netherlands (F.W.A., J.v.S.).,Department of Medical Genetics, Center for Molecular Medicine, University Medical Center Utrecht, University Utrecht, The Netherlands (P.I.W.d.B.).,Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University Utrecht, The Netherlands (P.I.W.d.B.).,Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy, University Medical Center Utrecht, University Utrecht, The Netherlands (G.P.).,Durrer Center for Cardiogenetic Research, Netherlands Heart Institute, Utrecht (F.W.A.).,Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom (F.W.A.).,Institute of Health Informatics, University College London, London, United Kingdom (F.W.A.)
| | - Folkert W den Ruijter
- Department of Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, University Utrecht, The Netherlands (F.W.A., J.v.S.)
| | - Paul I W de Bakker
- Department of Medical Genetics, Center for Molecular Medicine, University Medical Center Utrecht, University Utrecht, The Netherlands (P.I.W.d.B.).,Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University Utrecht, The Netherlands (P.I.W.d.B.)
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, Division Heart and Lungs, University Medical Center Utrecht, University Utrecht, The Netherlands (S.W.v.d.L., M.A.S., S.H., H.M.d.R., G.P.).,Department of Clinical Genetics, University Medical Center Utrecht, University Utrecht, The Netherlands (M.A.S.).,Laboratory of Clinical Chemistry and Hematology, Division Laboratories and Pharmacy, University Medical Center Utrecht, University Utrecht, The Netherlands (G.P.)
| |
Collapse
|
34
|
Durham AL, Speer MY, Scatena M, Giachelli CM, Shanahan CM. Role of smooth muscle cells in vascular calcification: implications in atherosclerosis and arterial stiffness. Cardiovasc Res 2019. [PMID: 29514202 PMCID: PMC5852633 DOI: 10.1093/cvr/cvy010] [Citation(s) in RCA: 726] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Vascular calcification is associated with a significant increase in all-cause mortality and atherosclerotic plaque rupture. Calcification has been determined to be an active process driven in part by vascular smooth muscle cell (VSMC) transdifferentiation within the vascular wall. Historically, VSMC phenotype switching has been viewed as binary, with the cells able to adopt a physiological contractile phenotype or an alternate ‘synthetic’ phenotype in response to injury. More recent work, including lineage tracing has however revealed that VSMCs are able to adopt a number of phenotypes, including calcific (osteogenic, chondrocytic, and osteoclastic), adipogenic, and macrophagic phenotypes. Whilst the mechanisms that drive VSMC differentiation are still being elucidated it is becoming clear that medial calcification may differ in several ways from the intimal calcification seen in atherosclerotic lesions, including risk factors and specific drivers for VSMC phenotype changes and calcification. This article aims to compare and contrast the role of VSMCs in driving calcification in both atherosclerosis and in the vessel media focusing on the major drivers of calcification, including aging, uraemia, mechanical stress, oxidative stress, and inflammation. The review also discusses novel findings that have also brought attention to specific pro- and anti-calcifying proteins, extracellular vesicles, mitochondrial dysfunction, and a uraemic milieu as major determinants of vascular calcification.
Collapse
Affiliation(s)
- Andrew L Durham
- Division of Cardiology, James Black Centre, Kings College London, Denmark Hill, London, SE5 9NU, UK
| | - Mei Y Speer
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Marta Scatena
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Cecilia M Giachelli
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Catherine M Shanahan
- Division of Cardiology, James Black Centre, Kings College London, Denmark Hill, London, SE5 9NU, UK
| |
Collapse
|
35
|
Sclerostin as Regulatory Molecule in Vascular Media Calcification and the Bone-Vascular Axis. Toxins (Basel) 2019; 11:toxins11070428. [PMID: 31330917 PMCID: PMC6669501 DOI: 10.3390/toxins11070428] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 01/18/2023] Open
Abstract
Sclerostin is a well-known inhibitor of bone formation that acts on Wnt/β-catenin signaling. This manuscript considers the possible role of sclerostin in vascular calcification, a process that shares many similarities with physiological bone formation. Rats were exposed to a warfarin-containing diet to induce vascular calcification. Vascular smooth muscle cell transdifferentiation, vascular calcification grade, and bone histomorphometry were examined. The presence and/or production of sclerostin was investigated in serum, aorta, and bone. Calcified human aortas were investigated to substantiate clinical relevance. Warfarin-exposed rats developed vascular calcifications in a time-dependent manner which went along with a progressive increase in serum sclerostin levels. Both osteogenic and adipogenic pathways were upregulated in calcifying vascular smooth muscle cells, as well as sclerostin mRNA and protein levels. Evidence for the local vascular action of sclerostin was found both in human and rat calcified aortas. Warfarin exposure led to a mildly decreased bone and mineralized areas. Osseous sclerostin production and bone turnover did not change significantly. This study showed local production of sclerostin in calcified vessels, which may indicate a negative feedback mechanism to prevent further calcification. Furthermore, increased levels of serum sclerostin, probably originating from excessive local production in calcified vessels, may contribute to the linkage between vascular pathology and impaired bone mineralization.
Collapse
|
36
|
Davies OG, Cox SC, Azoidis I, McGuinness AJA, Cooke M, Heaney LM, Davis ET, Jones SW, Grover LM. Osteoblast-Derived Vesicle Protein Content Is Temporally Regulated During Osteogenesis: Implications for Regenerative Therapies. Front Bioeng Biotechnol 2019; 7:92. [PMID: 31119130 PMCID: PMC6504811 DOI: 10.3389/fbioe.2019.00092] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/12/2019] [Indexed: 02/02/2023] Open
Abstract
Osteoblast-derived extracellular vesicles (EV) are a collection of secreted (sEVs) and matrix-bound nanoparticles that function as foci for mineral nucleation and accumulation. Due to the fact sEVs can be isolated directly from the culture medium of mineralizing osteoblasts, there is growing interest their application regenerative medicine. However, at present therapeutic advancements are hindered by a lack of understanding of their precise temporal contribution to matrix mineralization. This study advances current knowledge by temporally aligning sEV profile and protein content with mineralization status. sEVs were isolated from mineralizing primary osteoblasts over a period of 1, 2, and 3 weeks. Bimodal particle distributions were observed (weeks 1 and 3: 44 and 164 nm; week 2: 59 and 220 nm), indicating a heterogeneous population with dimensions characteristic of exosome- (44 and 59 nm) and microvesicle-like (164 and 220 nm) particles. Proteomic characterization by liquid chromatography tandem-mass spectrometry (LC-MS/MS) revealed a declining correlation in EV-localized proteins as mineralization advanced, with Pearson correlation-coefficients of 0.79 (week 1 vs. 2), 0.6 (2 vs. 3) and 0.46 (1 vs. 3), respectively. Principal component analysis (PCA) further highlighted a time-dependent divergence in protein content as mineralization advanced. The most significant variations were observed at week 3, with a significant (p < 0.05) decline in particle concentration, visual evidence of EV rupture and enhanced mineralization. A total of 116 vesicle-localized proteins were significantly upregulated at week 3 (56% non-specifically, 19% relative to week 1, 25% relative to week 2). Gene ontology enrichment analysis of these proteins highlighted overrepresentation of genes associated with matrix organization. Of note, increased presence of phospholipid-binding and calcium channeling annexin proteins (A2, A5, and A6) indicative of progressive variations in the nucleational capacity of vesicles, as well as interaction with the surrounding ECM. We demonstrate sEV-mediated mineralization is dynamic process with variations in vesicle morphology and protein content having a potential influence on developmental changes matrix organization. These findings have implications for the selection and application of EVs for regenerative applications.
Collapse
Affiliation(s)
- Owen G. Davies
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Sophie C. Cox
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Ioannis Azoidis
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Adam J. A. McGuinness
- Physical Sciences for Health Doctoral Training Centre, University of Birmingham, Birmingham, United Kingdom
| | - Megan Cooke
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
- Physical Sciences for Health Doctoral Training Centre, University of Birmingham, Birmingham, United Kingdom
| | - Liam M. Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | | | - Simon W. Jones
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Liam M. Grover
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
37
|
Kukida M, Mogi M, Kan-no H, Tsukuda K, Bai HY, Shan BS, Yamauchi T, Higaki A, Min LJ, Iwanami J, Okura T, Higaki J, Horiuchi M. AT2 receptor stimulation inhibits phosphate-induced vascular calcification. Kidney Int 2019; 95:138-148. [DOI: 10.1016/j.kint.2018.07.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 07/09/2018] [Accepted: 07/26/2018] [Indexed: 02/06/2023]
|
38
|
Gilham D, Tsujikawa LM, Sarsons CD, Halliday C, Wasiak S, Stotz SC, Jahagirdar R, Sweeney M, Johansson JO, Wong NCW, Kalantar-Zadeh K, Kulikowski E. Apabetalone downregulates factors and pathways associated with vascular calcification. Atherosclerosis 2018; 280:75-84. [PMID: 30476723 DOI: 10.1016/j.atherosclerosis.2018.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/28/2018] [Accepted: 11/07/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS Apabetalone is an inhibitor of bromodomain and extraterminal (BET) proteins. In clinical trials, apabetalone reduced the incidence of major adverse cardiac events (MACE) in patients with cardiovascular disease and reduced circulating factors that promote vascular calcification (VC). Because VC contributes to MACE, effects of apabetalone on pro-calcific processes were examined. METHODS AND RESULTS Apabetalone inhibited extracellular calcium deposition and opposed induction of transdifferentiation markers in human coronary artery vascular smooth muscle cells (VSMCs) under osteogenic culture conditions. Tissue-nonspecific alkaline phosphatase (TNAP) is a key contributor to VC, and apabetalone suppressed osteogenic induction of the mRNA, protein and enzyme activity. The liver is a major source of circulating TNAP, and apabetalone also downregulated TNAP expression in primary human hepatocytes. BRD4, a transcriptional regulator and target of apabetalone, has been linked to calcification. Osteogenic transdifferentiation of VSMCs resulted in disassembly of 100 BRD4-rich enhancers, with concomitant enlargement of remaining enhancers. Apabetalone reduced the size of BRD4-rich enhancers, consistent with disrupting BRD4 association with chromatin. 38 genes were uniquely associated with BRD4-rich enhancers in osteogenic conditions; 11 were previously associated with calcification. Apabetalone reduced levels of BRD4 on many of these enhancers, which correlated with decreased expression of the associated gene. Bioinformatics revealed BRD4 may cooperate with 7 specific transcription factors to promote transdifferentiation and calcification. CONCLUSIONS Apabetalone counters transdifferentiation and calcification of VSMCs via an epigenetic mechanism involving specific transcription factors. The mechanistic findings, combined with evidence from clinical trials, support further development of apabetalone as a therapeutic for VC.
Collapse
|
39
|
Chellan B, Sutton NR, Hofmann Bowman MA. S100/RAGE-Mediated Inflammation and Modified Cholesterol Lipoproteins as Mediators of Osteoblastic Differentiation of Vascular Smooth Muscle Cells. Front Cardiovasc Med 2018; 5:163. [PMID: 30467547 PMCID: PMC6235906 DOI: 10.3389/fcvm.2018.00163] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/19/2018] [Indexed: 01/01/2023] Open
Abstract
Arterial calcification is a feature of atherosclerosis and shares many risk factors including diabetes, dyslipidemia, chronic kidney disease, hypertension, and age. Although there is overlap in risk factors, anti-atherosclerotic therapies, including statins, fail to reduce arterial, and aortic valve calcifications. This suggests that low density lipoprotein (LDL) may not be the main driver for aortic valve disease and arterial calcification. This review focuses on modified LDLs and their role in mediating foam cell formation in smooth muscle cells (SMCs), with special emphasis on enzyme modified non-oxidized LDL (ELDL). In vivo, ELDL represents one of the many forms of modified LDLs present in the atherosclerotic vessel. Phenotypic changes of macrophages and SMCs brought about by the uptake of modified LDLs overlap significantly in an atherosclerotic milieu, making it practically impossible to differentiate between the effects from oxidized LDL, ELDL, and other LDL modification. By studying in vitro-generated modifications of LDL, we were able to demonstrate marked differences in the transcriptome of human coronary artery SMCs (HCASMCs) upon uptake of ELDL, OxLDL, and native LDL, indicating that specific modifications of LDL in atherosclerotic plaques may determine the biology and functional consequences in vasculature. Enzyme-modified non-oxidized LDL (ELDL) induces calcification of SMCs and this is associated with reduced mRNA levels for genes protective for calcification (ENPP1, MGP) and upregulation of osteoblastic genes. A second focus of this review is on the synergy between hyperlipidemia and accelerated calcification In vivo in a mouse models with transgenic expression of human S100A12. We summarize mechanisms of S100A12/RAGE mediated vascular inflammation promoting vascular and valve calcification in vivo.
Collapse
Affiliation(s)
- Bijoy Chellan
- Department of Medicine, University of Illinois, Chicago, IL, United States
| | - Nadia R Sutton
- Department of Medicine, University of Michigan, Ann Arbor, MI, United States
| | | |
Collapse
|
40
|
Wang X, Zhang X, Li F, Ji Q. MiR‐128‐3p accelerates cardiovascular calcification and insulin resistance through ISL1‐dependent Wnt pathway in type 2 diabetes mellitus rats. J Cell Physiol 2018; 234:4997-5010. [PMID: 30341898 DOI: 10.1002/jcp.27300] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 08/01/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Xin‐Yong Wang
- Department of Internal Medicine Linyi Jiaotong Hospital Linyi China
| | - Xian‐Zhao Zhang
- Department of Cardiology Linyi People's Hospital Linyi China
| | - Feng Li
- Clinical Laboratory The Third People's Hospital of Linyi Linyi China
| | - Qing‐Rong Ji
- Department of Cardiology Linyi People's Hospital Linyi China
| |
Collapse
|
41
|
Shen ZX, Yang QZ, Li C, Du LJ, Sun XN, Liu Y, Sun JY, Gu HH, Sun YM, Wang J, Duan SZ. Myeloid peroxisome proliferator-activated receptor gamma deficiency aggravates myocardial infarction in mice. Atherosclerosis 2018; 274:199-205. [PMID: 29800789 DOI: 10.1016/j.atherosclerosis.2018.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 10/17/2022]
Abstract
BACKGROUND AND AIMS Agonists of peroxisome proliferator-activated receptor gamma (Pparγ) have been demonstrated to reduce the risk of myocardial infarction (MI) in clinical trials and animal experiments. However, the cellular and molecular mechanisms are not completely understood. We aimed to reveal the functions of myeloid Pparγ in MI and explore the potential mechanisms in this study. METHODS Myeloid Pparγ knockout (MPGKO) mice (n = 12) and control mice (n = 8) underwent coronary artery ligation to induce MI. Another cohort of MPGKO mice and control mice underwent coronary artery ligation and were then treated with IgG or neutralizing antibodies against interleukin (IL)-1β. Infarct size was determined by TTC staining and cardiac function was measured using echocardiography. Conditioned media from GW9662- or vehicle-treated macrophages were used to treat H9C2 cardiomyocyte cell line. Gene expression was analyzed using quantitative PCR. Reactive oxygen species were measured using flow cytometry. RESULTS Myeloid Pparγ deficiency significantly increased myocardial infarct size. Cardiac hypertrophy was also exacerbated in MPGKO mice, with upregulation of β-myosin heavy chain (Mhc) and brain natriuretic peptide (Bnp) and downregulation of α-Mhc in the non-infarcted zone. Conditioned media from GW9662-treated macrophages increased expression of β-Mhc and Bnp in H9C2 cells. Echocardiographic measurements showed that MPGKO mice had worsen cardiac dysfunction after MI. Myeloid Pparγ deficiency increased gene expression of NADPH oxidase subunits (Nox2 and Nox4) in the non-infarcted zone after MI. Conditioned media from GW9662-treated macrophages increased reactive oxygen species in H9C2 cells. Expression of inflammatory genes such as IL-1β and IL-6 was upregulated in the non-infarcted zone of MPGKO mice after MI. With the injection of neutralizing antibodies against IL-1β, control mice and MPGKO mice had comparable cardiac function and expression of inflammatory genes after MI. CONCLUSIONS Myeloid Pparγ deficiency exacerbates MI, likely through increased oxidative stress and cardiac inflammation.
Collapse
Affiliation(s)
- Zhu-Xia Shen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, 200040, China
| | - Qing-Zhen Yang
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Chao Li
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lin-Juan Du
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xue-Nan Sun
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China; Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yan Liu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Jian-Yong Sun
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
| | - Hui-Hui Gu
- Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, 200040, China
| | - Yu-Min Sun
- Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, 200040, China
| | - Jun Wang
- Department of Cardiology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, 200040, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Ninth People's Hospital, School of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China.
| |
Collapse
|
42
|
Ramachandran B, Stabley JN, Cheng SL, Behrmann AS, Gay A, Li L, Mead M, Kozlitina J, Lemoff A, Mirzaei H, Chen Z, Towler DA. A GTPase-activating protein-binding protein (G3BP1)/antiviral protein relay conveys arteriosclerotic Wnt signals in aortic smooth muscle cells. J Biol Chem 2018; 293:7942-7968. [PMID: 29626090 DOI: 10.1074/jbc.ra118.002046] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/13/2018] [Indexed: 12/21/2022] Open
Abstract
In aortic vascular smooth muscle (VSM), the canonical Wnt receptor LRP6 inhibits protein arginine (Arg) methylation, a new component of noncanonical Wnt signaling that stimulates nuclear factor of activated T cells (viz NFATc4). To better understand how methylation mediates these actions, MS was performed on VSM cell extracts from control and LRP6-deficient mice. LRP6-dependent Arg methylation was regulated on >500 proteins; only 21 exhibited increased monomethylation (MMA) with concomitant reductions in dimethylation. G3BP1, a known regulator of arteriosclerosis, exhibited a >30-fold increase in MMA in its C-terminal domain. Co-transfection studies confirm that G3BP1 (G3BP is Ras-GAP SH3 domain-binding protein) methylation is inhibited by LRP6 and that G3BP1 stimulates NFATc4 transcription. NFATc4 association with VSM osteopontin (OPN) and alkaline phosphatase (TNAP) chromatin was increased with LRP6 deficiency and reduced with G3BP1 deficiency. G3BP1 activation of NFATc4 mapped to G3BP1 domains supporting interactions with RIG-I (retinoic acid inducible gene I), a stimulus for mitochondrial antiviral signaling (MAVS) that drives cardiovascular calcification in humans when mutated in Singleton-Merten syndrome (SGMRT2). Gain-of-function SGMRT2/RIG-I mutants increased G3BP1 methylation and synergized with osteogenic transcription factors (Runx2 and NFATc4). A chemical antagonist of G3BP, C108 (C108 is 2-hydroxybenzoic acid, 2-[1-(2-hydroxyphenyl)ethylidene]hydrazide CAS 15533-09-2), down-regulated RIG-I-stimulated G3BP1 methylation, Wnt/NFAT signaling, VSM TNAP activity, and calcification. G3BP1 deficiency reduced RIG-I protein levels and VSM osteogenic programs. Like G3BP1 and RIG-I deficiency, MAVS deficiency reduced VSM osteogenic signals, including TNAP activity and Wnt5-dependent nuclear NFATc4 levels. Aortic calcium accumulation is decreased in MAVS-deficient LDLR-/- mice fed arteriosclerotic diets. The G3BP1/RIG-I/MAVS relay is a component of Wnt signaling. Targeting this relay may help mitigate arteriosclerosis.
Collapse
Affiliation(s)
- Bindu Ramachandran
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - John N Stabley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Su-Li Cheng
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Abraham S Behrmann
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Austin Gay
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Li Li
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Megan Mead
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Julia Kozlitina
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Hamid Mirzaei
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Zhijian Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Dwight A Towler
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390.
| |
Collapse
|
43
|
Loss of the adaptor protein ShcA in endothelial cells protects against monocyte macrophage adhesion, LDL-oxydation, and atherosclerotic lesion formation. Sci Rep 2018. [PMID: 29540796 PMCID: PMC5852050 DOI: 10.1038/s41598-018-22819-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
ShcA is an adaptor protein that binds to the cytoplasmic tail of receptor tyrosine kinases and of the Low Density Lipoprotein-related receptor 1 (LRP1), a trans-membrane receptor that protects against atherosclerosis. Here, we examined the role of endothelial ShcA in atherosclerotic lesion formation. We found that atherosclerosis progression was markedly attenuated in mice deleted for ShcA in endothelial cells, that macrophage content was reduced at the sites of lesions, and that adhesion molecules such as the intercellular adhesion molecule-1 (ICAM-1) were severely reduced. Our data indicate that transcriptional regulation of ShcA by the zinc-finger E-box-binding homeobox 1 (ZEB1) and the Hippo pathway effector YAP, promotes ICAM-1 expression independently of p-NF-κB, the primary driver of adhesion molecules expressions. In addition, ShcA suppresses endothelial Akt and nitric oxide synthase (eNOS) expressions. Thus, through down regulation of eNOS and ZEB1-mediated ICAM-1 up regulation, endothelial ShcA promotes monocyte-macrophage adhesion and atherosclerotic lesion formation. Reducing ShcA expression in endothelial cells may represent an obvious therapeutic approach to prevent atherosclerosis.
Collapse
|
44
|
Shao Y, Zheng Q, Wang W, Xin N, Song X, Zhao C. Biological functions of macrophage-derived Wnt5a, and its roles in human diseases. Oncotarget 2018; 7:67674-67684. [PMID: 27608847 PMCID: PMC5341904 DOI: 10.18632/oncotarget.11874] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 08/27/2016] [Indexed: 12/31/2022] Open
Abstract
Wnt5a is implicated in development and tissue homeostasis by activating β-catenin-independent pathway. Excessive production of Wnt5a is related to some human diseases. Macrophage recruitment is a character of inflammation and cancer, therefore macrophage-derived Wnt5a is supposed to be a player in these conditions. Actually, macrophage-derived Wnt5a maintains macrophage immune function, stimulates pro-inflammatory cytokine release, and induces angiogenesis and lymphangiogenesis. Furthermore, macrophage-derived Wnt5a is involved in insulin resistance, atherosclerosis and cancer. These findings indicate that macrophage-derived Wnt5a may be a target in the treatment of these diseases. Notably, unlike macrophages, the exact role of macrophage-derived Wnt5a in bacterial infection remains largely unknown.
Collapse
Affiliation(s)
- Yue Shao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Qianqian Zheng
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Na Xin
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Xiaowen Song
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Chenghai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
45
|
Liu L, Liu Y, Zhang Y, Bi X, Nie L, Liu C, Xiong J, He T, Xu X, Yu Y, Yang K, Gu J, Huang Y, Zhang J, Zhang Z, Zhang B, Zhao J. High phosphate-induced downregulation of PPARγ contributes to CKD-associated vascular calcification. J Mol Cell Cardiol 2017; 114:264-275. [PMID: 29197521 DOI: 10.1016/j.yjmcc.2017.11.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 02/04/2023]
Abstract
Medial arterial calcification associated with hyperphosphatemia is a main cause of cardiovascular mortality in patients with chronic kidney disease (CKD), but the mechanisms underlying high phosphate-induced vascular calcification remain largely unknown. Here, we observed a significant decrease in the expression of peroxisome proliferator-activated receptor-gamma (PPARγ) in calcified arteries both in CKD patients and in a mouse model of CKD with hyperphosphatemia. In vitro, high phosphate treatment led to a decreased expression of PPARγ in mouse vascular smooth muscle cells (VMSCs), accompanied by apparent osteogenic differentiation and calcification. Pretreatment with PPARγ agonist rosiglitazone significantly reversed high phosphate-induced VSMCs calcification. Further investigation showed that methyl-CpG binding protein 2 (Mecp2)-mediated epigenetic repression was involved in high phosphate-induced PPARγ downregulation. Moreover, the expression of Klotho that has the ability to inhibit vascular calcification by regulating phosphate uptake decreased with the PPARγ reduction in VSMCs after high phosphate treatment, and rosiglitazone failed to inhibit high phosphate-induced calcification in VSMCs with knockdown of Klotho or in aortic rings from Klotho-deficient (kl/kl) mice. Finally, an in vivo study demonstrated that oral administration of rosiglitazone could increase Klotho expression and protect against high phosphate-induced vascular calcification in CKD mice. These findings suggest that the inhibition of PPARγ expression may contribute to the pathogenesis of high phosphate-induced vascular calcification, which may provide a new therapeutic target for vascular calcification in CKD patients.
Collapse
Affiliation(s)
- Liang Liu
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Yong Liu
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Ying Zhang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Xianjin Bi
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Ling Nie
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Chi Liu
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Jiachuan Xiong
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Ting He
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Xinlin Xu
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Yanlin Yu
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Ke Yang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Jun Gu
- State Key Laboratory of Protein and Plant Gene Research, College of Life Science, Peking University, Beijing, PR China
| | - Yunjian Huang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Jingbo Zhang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Zhiren Zhang
- Department of Basic Medicine, Institute of Immunology, Third Military Medical University, Chongqing, PR China
| | - Bo Zhang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Jinghong Zhao
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China.
| |
Collapse
|
46
|
Xian X, Ding Y, Dieckmann M, Zhou L, Plattner F, Liu M, Parks JS, Hammer RE, Boucher P, Tsai S, Herz J. LRP1 integrates murine macrophage cholesterol homeostasis and inflammatory responses in atherosclerosis. eLife 2017; 6:e29292. [PMID: 29144234 PMCID: PMC5690284 DOI: 10.7554/elife.29292] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 10/22/2017] [Indexed: 12/11/2022] Open
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) is a multifunctional cell surface receptor with diverse physiological roles, ranging from cellular uptake of lipoproteins and other cargo by endocytosis to sensor of the extracellular environment and integrator of a wide range of signaling mechanisms. As a chylomicron remnant receptor, LRP1 controls systemic lipid metabolism in concert with the LDL receptor in the liver, whereas in smooth muscle cells (SMC) LRP1 functions as a co-receptor for TGFβ and PDGFRβ in reverse cholesterol transport and the maintenance of vascular wall integrity. Here we used a knockin mouse model to uncover a novel atheroprotective role for LRP1 in macrophages where tyrosine phosphorylation of an NPxY motif in its intracellular domain initiates a signaling cascade along an LRP1/SHC1/PI3K/AKT/PPARγ/LXR axis to regulate and integrate cellular cholesterol homeostasis through the expression of the major cholesterol exporter ABCA1 with apoptotic cell removal and inflammatory responses.
Collapse
Affiliation(s)
- Xunde Xian
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
| | - Yinyuan Ding
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
- Key Laboratory of Medical Electrophysiology, Ministry of Education of ChinaInstitute of Cardiovascular Research, Southwest Medical UniversityLuzhouChina
| | - Marco Dieckmann
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
| | - Li Zhou
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
| | - Florian Plattner
- Department of PsychiatryUniversity of Texas Southwestern Medical CenterDallasUnited States
- Center for Translational Neurodegeneration ResearchUniversity of Texas Southwestern Medical CenterDallasUnited States
| | - Mingxia Liu
- Section on Molecular Medicine, Department of Internal MedicineWake Forest School of MedicineWinston-SalemNorth Carolina
| | - John S Parks
- Section on Molecular Medicine, Department of Internal MedicineWake Forest School of MedicineWinston-SalemNorth Carolina
| | - Robert E Hammer
- Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasUnited States
| | | | - Shirling Tsai
- Department of SurgeryUT Southwestern Medical CenterDallasUnited States
- Dallas VA Medical CenterDallasUnited States
| | - Joachim Herz
- Departments of Molecular GeneticsUT Southwestern Medical CenterDallasUnited States
- Center for Translational Neurodegeneration ResearchUniversity of Texas Southwestern Medical CenterDallasUnited States
- Department of NeuroscienceUT SouthwesternDallasUnited States
- Department of Neurology and NeurotherapeuticsUT SouthwesternDallasUnited States
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Cardiometabolic diseases increasingly afflict our aging, dysmetabolic population. Complex signals regulating low-density lipoprotein receptor-related protein (LRP) and frizzled protein family members - the plasma membrane receptors for the cadre of Wnt polypeptide morphogens - contribute to the control of cardiovascular homeostasis. RECENT FINDINGS Both canonical (β-catenin-dependent) and noncanonical (β-catenin-independent) Wnt signaling programs control vascular smooth muscle (VSM) cell phenotypic modulation in cardiometabolic disease. LRP6 limits VSM proliferation, reduces arteriosclerotic transcriptional reprogramming, and preserves insulin sensitivity while LRP5 restrains foam cell formation. Adipose, skeletal muscle, macrophages, and VSM have emerged as important sources of circulating Wnt ligands that are dynamically regulated during the prediabetes-diabetes transition with cardiometabolic consequences. Platelets release Dkk1, a LRP5/LRP6 inhibitor that induces endothelial inflammation and the prosclerotic endothelial-mesenchymal transition. By contrast, inhibitory secreted frizzled-related proteins shape the Wnt signaling milieu to limit myocardial inflammation with ischemia-reperfusion injury. VSM sclerostin, an inhibitor of canonical Wnt signaling in bone, restrains remodeling that predisposes to aneurysm formation, and is downregulated in aneurysmal vessels by epigenetic methylation. SUMMARY Components of the Wnt signaling cascade represent novel targets for pharmacological intervention in cardiometabolic disease. Conversely, strategies targeting the Wnt signaling cascade for other therapeutic purposes will have cardiovascular consequences that must be delineated to establish clinically useful pharmacokinetic-pharmacodynamic relationships.
Collapse
Affiliation(s)
- Austin Gay
- Department of Internal Medicine-Endocrine Division, UT Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
48
|
Gao M, Chen T, Wu L, Zhao X, Mao H, Xing C. Effect of pioglitazone on the calcification of rat vascular smooth muscle cells through the downregulation of the Wnt/β‑catenin signaling pathway. Mol Med Rep 2017; 16:6208-6213. [PMID: 28849074 DOI: 10.3892/mmr.2017.7308] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 07/03/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the effect and possible mechanism of pioglitazone (PIO) on the calcification of rat vascular smooth muscle cells (VSMCs) in vitro. β‑glycerophosphate (β‑GP; 10 mmol/l) was used to induce calcification of VSMCs treated with a range of concentrations (5, 10, 15 and 20 µmol/l) of PIO for 12 days. Calcium deposits were revealed by Alizarin red staining. Extracellular calcium content was detected using a calcium assay kit. Western blotting was used to measure the expression of α‑smooth muscle actin (α‑SMA), runt‑related transcription factor 2 (Runx2), bone morphogenetic protein‑2 (BMP2), β‑catenin, glycogen synthase kinase‑3β (GSK‑3β), phosphorylated (p)‑GSK‑3β and cyclin‑D1. A total of 10 mmol/l β‑GP, 20 µmol/l PIO and 20 µmol/l peroxisome proliferator‑activated receptor γ (PPAR γ) antagonist GW9662, was added to the cell culture media. The changes of the above indexes were observed. The calcium content in the calcification group, treated with high phosphorus, increased significantly compared with the controls (P<0.05) and all different concentrations of PIO reduced extracellular calcium content (P<0.05). Alizarin red staining was positive in calcified VSMCs and PIO (20 µmol/l) intervention group was almost negative. The expressions of Runx2, β‑catenin, p‑GSK‑3β, BMP2 and cyclin‑D1 increased significantly in the calcification group, and treatment with 20 µmol/l PIO downregulated the expression of all the above proteins, while upregulating the expression of α‑SMA. The PPAR γ antagonist GW9662 could partly inhibit the effect of PIO on calcified VSMCs. The results of the present study indicated that PIO can alleviate the calcification of rat aortic VSMCs induced by β‑GP via inhibiting the activity of the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Min Gao
- Department of Nephrology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Tianlei Chen
- Department of Nephrology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lin Wu
- Department of Nephrology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiufen Zhao
- Department of Nephrology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Huijuan Mao
- Department of Nephrology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Changying Xing
- Department of Nephrology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
49
|
Vongpromek R, Bos S, Ten Kate GJR, Bujo H, Jiang M, Nieman K, Schneider W, Roeters van Lennep JE, Verhoeven AJM, Sijbrands EJG, Mulder MT. Soluble LR11 associates with aortic root calcification in asymptomatic treated male patients with familial hypercholesterolemia. Atherosclerosis 2017. [PMID: 28637586 DOI: 10.1016/j.atherosclerosis.2017.06.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS Despite statin treatment, a high prevalence of severe vascular calcification is found in patients with familial hypercholesterolemia (FH). We assessed the relation between the circulating soluble form of low-density lipoprotein receptor relative with 11 ligand-binding repeats (sLR11), a risk factor for cardiovascular disease, and vascular calcification in asymptomatic statin-treated heterozygous FH patients. METHODS In 123 asymptomatic heterozygous FH patients (age 40-69 years), aortic root (ARC), aortic valve (AVC) and coronary artery calcification (CAC) were determined with CT-based calcium scoring expressed in Agatston units. Plasma sLR11 levels were measured by sandwich ELISA. RESULTS Seventy-three patients displayed ARC, 48 had AVC and 96 CAC. Plasma sLR11 levels were positively correlated with the presence of ARC (r = 0.2, p = 0.03), but not with AVC or CAC. The correlation between sLR11 levels and ARC was restricted to male FH patients (r = 0.31, p = 0.006). Multivariate logistic analyses showed that the association of plasma sLR11 with the presence of ARC was independent of other determinants (Adjusted Odds Ratio, 2.01 (95% CI = 1.28-3.16) p = 0.002). CONCLUSIONS Plasma sLR11 is associated with ARC in male FH patients and may be mechanistically involved in the differential distribution of atherosclerotic lesions in the vasculature.
Collapse
Affiliation(s)
- Ranitha Vongpromek
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sven Bos
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gert-Jan R Ten Kate
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Hideaki Bujo
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University, Sakura Medical Center, Sakura, Japan
| | - Meizi Jiang
- Department of Clinical-Laboratory and Experimental-Research Medicine, Toho University, Sakura Medical Center, Sakura, Japan
| | - Koen Nieman
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Radiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Wolfgang Schneider
- Department of Medical Biochemistry, Medical University of Vienna, Max. F. Perutz Laboratories, Vienna, Austria
| | - Jeanine E Roeters van Lennep
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Adrie J M Verhoeven
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Eric J G Sijbrands
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Laboratory of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
50
|
Towler DA. "Osteotropic" Wnt/LRP Signals: High-Wire Artists in a Balancing Act Regulating Aortic Structure and Function. Arterioscler Thromb Vasc Biol 2017; 37:392-395. [PMID: 28228445 PMCID: PMC5324723 DOI: 10.1161/atvbaha.116.308915] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Dwight A Towler
- From the Department of Internal Medicine, Endocrine Division, UT Southwestern Medical Center, Dallas, TX.
| |
Collapse
|