1
|
Morales-Sánchez P, Lambert C, Ares-Blanco J, Suárez-Gutiérrez L, Villa-Fernández E, Garcia AV, García-Villarino M, Tejedor JR, Fraga MF, Torre EM, Pujante P, Delgado E. Circulating miRNA expression in long-standing type 1 diabetes mellitus. Sci Rep 2023; 13:8611. [PMID: 37244952 DOI: 10.1038/s41598-023-35836-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
Type 1 diabetes is a chronic autoimmune disease which results in inefficient regulation of glucose homeostasis and can lead to different vascular comorbidities through life. In this study we aimed to analyse the circulating miRNA expression profile of patients with type 1 diabetes, and with no other associated pathology. For this, fasting plasma was obtained from 85 subjects. Next generation sequencing analysis was firstly performed to identify miRNAs that were differentially expressed between groups (20 patients vs. 10 controls). hsa-miR-1-3p, hsa-miR-200b-3p, hsa-miR-9-5p, and hsa-miR-1200 expression was also measured by Taqman RT-PCR to validate the observed changes (34 patients vs. 21 controls). Finally, through a bioinformatic approach, the main pathways affected by the target genes of these miRNAs were studied. Among the studied miRNAs, hsa-miR-1-3p expression was found significantly increased in patients with type 1 diabetes compared to controls, and positively correlated with glycated haemoglobin levels. Additionally, by using a bioinformatic approach, we could observe that changes in hsa-miR-1-3p directly affect genes involved in vascular development and cardiovascular pathologies. Our results suggest that, circulating hsa-miR-1-3p in plasma, together with glycaemic control, could be used as prognostic biomarkers in type 1 diabetes, helping to prevent the development of vascular complications in these patients.
Collapse
Affiliation(s)
- Paula Morales-Sánchez
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Lambert
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- University of Barcelona, Barcelona, Spain.
| | - Jessica Ares-Blanco
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
| | - Lorena Suárez-Gutiérrez
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Elsa Villa-Fernández
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Ana Victoria Garcia
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Miguel García-Villarino
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
| | - Juan Ramón Tejedor
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Oviedo, Asturias, Spain
- Institute of Oncology of Asturias (IUOPA), Oviedo, Asturias, Spain
- Department of Organisms and Systems Biology (B.O.S), University of Oviedo, Oviedo, Asturias, Spain
| | - Mario F Fraga
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), Oviedo, Asturias, Spain
- Institute of Oncology of Asturias (IUOPA), Oviedo, Asturias, Spain
- Department of Organisms and Systems Biology (B.O.S), University of Oviedo, Oviedo, Asturias, Spain
| | - Edelmiro Menéndez Torre
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
| | - Pedro Pujante
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain.
| | - Elías Delgado
- Endocrinology, Nutrition, Diabetes and Obesity Group (ENDO), Health Research Institute of the Principality of Asturias (ISPA), Av. Hospital Universitario s/n, 33011, Oviedo, Asturias, Spain.
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.
- Endocrinology and Nutrition Department, Asturias Central University Hospital, Oviedo, Asturias, Spain.
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain.
| |
Collapse
|
2
|
Wang J, Tian X, Yan C, Wu H, Bu Y, Li J, Liu D, Han Y. TCF7L1 Accelerates Smooth Muscle Cell Phenotypic Switching and Aggravates Abdominal Aortic Aneurysms. JACC Basic Transl Sci 2023; 8:155-170. [PMID: 36908661 PMCID: PMC9998605 DOI: 10.1016/j.jacbts.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 12/02/2022]
Abstract
Phenotypic switching of vascular smooth muscle cells is a central process in abdominal aortic aneurysm (AAA) pathology. We found that knockdown TCF7L1 (transcription factor 7-like 1), a member of the TCF/LEF (T cell factor/lymphoid enhancer factor) family of transcription factors, inhibits vascular smooth muscle cell differentiation. This study hints at potential interventions to maintain a normal, differentiated smooth muscle cell state, thereby eliminating the pathogenesis of AAA. In addition, our study provides insights into the potential use of TCF7L1 as a biomarker for AAA.
Collapse
Key Words
- AAA, abdominal aortic aneurysm
- AAV, adeno-associated virus
- Ang II, angiotensin II
- CVF, collagen volume fraction
- MMP, matrix metalloproteinase
- PBS, phosphate-buffered saline
- PCR, polymerase chain reaction
- SM22α, smooth muscle protein 22-α
- SMA, smooth muscle actin
- SRF, serum response factor
- TCF7L1
- TCF7L1, transcription factor 7-like 1
- VSMC, vascular smooth muscle cell
- abdominal aortic aneurysms
- cDNA, complementary DNA
- mRNA, messenger RNA
- phenotypic switching
- siRNA, small interfering RNA
- smooth muscle cell
Collapse
Affiliation(s)
- Jing Wang
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaoxiang Tian
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chenghui Yan
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Hanlin Wu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yuxin Bu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jia Li
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Dan Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
3
|
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: 57] [Impact Index Per Article: 19.0] [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
|
4
|
New Insights to the Crosstalk between Vascular and Bone Tissue in Chronic Kidney Disease-Mineral and Bone Disorder. Metabolites 2021; 11:metabo11120849. [PMID: 34940607 PMCID: PMC8708186 DOI: 10.3390/metabo11120849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022] Open
Abstract
Vasculature plays a key role in bone development and the maintenance of bone tissue throughout life. The two organ systems are not only linked in normal physiology, but also in pathophysiological conditions. The chronic kidney disease–mineral and bone disorder (CKD-MBD) is still the most serious complication to CKD, resulting in increased morbidity and mortality. Current treatment therapies aimed at the phosphate retention and parathyroid hormone disturbances fail to reduce the high cardiovascular mortality in CKD patients, underlining the importance of other factors in the complex syndrome. This review will focus on vascular disease and its interplay with bone disorders in CKD. It will present the very late data showing a direct effect of vascular calcification on bone metabolism, indicating a vascular-bone tissue crosstalk in CKD. The calcified vasculature not only suffers from the systemic effects of CKD but seems to be an active player in the CKD-MBD syndrome impairing bone metabolism and might be a novel target for treatment and prevention.
Collapse
|
5
|
Association of Circulating miR-145-5p and miR-let7c and Atherosclerotic Plaques in Hypertensive Patients. Biomolecules 2021; 11:biom11121840. [PMID: 34944484 PMCID: PMC8699419 DOI: 10.3390/biom11121840] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/16/2022] Open
Abstract
Aim: Hypertension is a strong risk factor for atherosclerosis. Increased carotid intima-media thickness (cIMT) and carotid plaques are considered subclinical markers of atherosclerosis. This study aimed at evaluating the serum expression of miRNAs previously related to adverse vascular remodeling and correlating them with carotid plaques and cIMT in hypertensive patients. Methods: We cross-sectionally evaluated the clinical and carotid characteristics as well as serum expression of miR-145-5p, miR-let7c, miR-92a, miR-30a and miR-451 in 177 hypertensive patients. Carotid plaques and cIMT were evaluated by ultrasound, and the expression of selected miRNAs was evaluated by a quantitative polymerase chain reaction. Results: Among all participants (age = 60.6 ± 10.7 years, 43% males), there were 59% with carotid plaques. We observed an increased expression of miR-145-5p (Fold Change = 2.0, p = 0.035) and miR-let7c (Fold Change = 3.8, p = 0.045) in participants with atherosclerotic plaque when compared to those without plaque. In the logistic regression analysis adjusted for relevant covariates, these miRNAs showed a stronger association with carotid plaques (miR-145-5p: Beta ± SE = 0.050 ± 0.020, p = 0.016 and miR-let7c: Beta ± SE = 0.056 ± 0.019, p = 0.003). Conclusions: Hypertensive patients with carotid plaques have an increased expression of miR-145-5p and miR-let7c, suggesting a potential role of these miRNAs as a biomarker for subclinical atherosclerosis in hypertensive individuals.
Collapse
|
6
|
Zhu Y, Ji JJ, Wang XD, Sun XJ, Li M, Wei Q, Ren LQ, Liu NF. Periostin promotes arterial calcification through PPARγ-related glucose metabolism reprogramming. Am J Physiol Heart Circ Physiol 2021; 320:H2222-H2239. [PMID: 33834866 DOI: 10.1152/ajpheart.01009.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Extracellular matrix (ECM) exerts a series of biological functions and contributes to almost 30% of the osteogenic process. Periostin is a secreted protein that can alter ECM remodeling in response to vascular injury. However, the role of periostin in vascular calcification has yet to be fully investigated. As found in this study, recombinant periostin accelerated the thoracic aortas calcification, increased the expression of glycolysis key enzymes, and disturbed the normal oxidative phosphorylation (OXPHOS) ex vivo, which could be alleviated by the peroxisome proliferation-activated receptor γ (PPARγ) agonist pioglitazone. In vascular smooth muscle cells (VSMCs), periostin promoted VSMC-osteoblastic phenotype transition and calcium deposition and suppressed PPARγ expression. Mechanistically, periostin caused overactivation of glycolysis and mitochondrial dysfunction in VSMCs as assessed by extracellular acidification rate, oxygen consumption rate, and mitochondrial respiratory chain complex activities. Targeted glycolysis inhibitors reduced mitochondrial calcium overload, apoptosis, and periostin-induced VSMCs calcification. PPARγ agonists preserved glycolysis and OXPHOS in the stimulated microenvironment and reversed periostin-promoted VSMC calcification. Furthermore, plasma periostin, lactate, and matrix Gla protein levels were measured in 274 patients undergoing computed tomography to determine coronary artery calcium score (Agatston score). Plasma periostin and lactate levels were both linked to an Agatston score in patients with coronary artery calcification (CAC). There was also a positive correlation between plasma periostin and lactate levels. This study suggests that downregulation of PPARγ is involved in the mechanism by which periostin accelerates arterial calcification partly through excessive glycolysis activation and unbalanced mitochondrial homeostasis.NEW & NOTEWORTHY Periostin caused arterial calcification, overactivated glycolysis, and damaged OXPHOS. PPARγ agonists alleviated periostin-promoted arterial calcification and corrected abnormal glycolysis and unbalanced mitochondrial homeostasis. There exists a relationship between periostin and lactate in patients with CAC.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Cardiology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, People's Republic of China
| | - Jing-Jing Ji
- Department of Cardiology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, People's Republic of China
| | - Xiao-Dong Wang
- Department of Cardiology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, People's Republic of China
| | - Xue-Jiao Sun
- Department of Cardiology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, People's Republic of China
| | - Min Li
- Department of Cardiology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, People's Republic of China
| | - Qin Wei
- Department of Cardiology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, People's Republic of China
| | - Li-Qun Ren
- Department of Geriatrics, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, People's Republic of China
| | - Nai-Feng Liu
- Department of Cardiology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
7
|
Donadon M, Santoro MM. The origin and mechanisms of smooth muscle cell development in vertebrates. Development 2021; 148:148/7/dev197384. [PMID: 33789914 DOI: 10.1242/dev.197384] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Smooth muscle cells (SMCs) represent a major structural and functional component of many organs during embryonic development and adulthood. These cells are a crucial component of vertebrate structure and physiology, and an updated overview of the developmental and functional process of smooth muscle during organogenesis is desirable. Here, we describe the developmental origin of SMCs within different tissues by comparing their specification and differentiation with other organs, including the cardiovascular, respiratory and intestinal systems. We then discuss the instructive roles of smooth muscle in the development of such organs through signaling and mechanical feedback mechanisms. By understanding SMC development, we hope to advance therapeutic approaches related to tissue regeneration and other smooth muscle-related diseases.
Collapse
Affiliation(s)
- Michael Donadon
- Department of Biology, University of Padua, Via U. Bassi 58B, 35121 Padua, Italy
| | - Massimo M Santoro
- Department of Biology, University of Padua, Via U. Bassi 58B, 35121 Padua, Italy
| |
Collapse
|
8
|
Zhao XY, Hu SY, Yang JL, Chen XM, Huang XL, Tang LJ, Gu L, Su L. A 3' Untranslated Region Polymorphism of CTNNB1 (Rs2953) Alters MiR-3161 Binding and Affects the Risk of Ischemic Stroke and Coronary Artery Disease in Chinese Han Population. Eur Neurol 2021; 84:85-95. [PMID: 33789307 DOI: 10.1159/000514543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 12/31/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND CTNNB1 is reported to be related to the pathological process of ischemic stroke (IS) and coronary artery disease (CAD). Polymorphism located in the 3' untranslated region (3'UTR) of a gene might affect gene expression by modifying binding sites for microRNAs (miRNAs). This study aimed to analyze the association between polymorphism rs2953, which locates in the 3'UTR of CTNNB1, and the risk of IS and CAD. METHODS The CTNNB1 messenger RNA (mRNA) expression level in peripheral venous blood was measured. In total, 533 patients with IS, 500 patients with CAD, and 531 healthy individuals were genotyped by Sequenom Mass-Array technology. The binding of miR-3161 to CTNNB1 was determined by dual-luciferase reporter assay. RESULTS The CTNNB1 mRNA expression level for the IS group was significantly lower than that for the control group. Rs2953 was significantly associated with both IS risk and CAD risk. Significant association was also found between polymorphism rs2953 and many conventional factors, such as serum lipid level, blood coagulation markers, blood glucose level, and homocysteine level in patients. Rs2953 T allele introduced a binding site to miRNA-3161 and thus decreased luciferase activity. CONCLUSION Polymorphism rs2953 is associated with the risk of both IS and CAD. Moreover, polymorphism rs2953 (T) introduces a binding site to miRNA-3161 and thus decreases luciferase activity in cell lines.
Collapse
Affiliation(s)
- Xin-Yi Zhao
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Shu-Yan Hu
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Jia-Lei Yang
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Xing-Mei Chen
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Xian-Li Huang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Lue-Jun Tang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Lian Gu
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Li Su
- School of Public Health, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, China
| |
Collapse
|
9
|
Klingenschmid G, Tschiderer L, Himmler G, Rungger G, Brugger S, Santer P, Willeit J, Kiechl S, Willeit P. Associations of Serum Dickkopf-1 and Sclerostin With Cardiovascular Events: Results From the Prospective Bruneck Study. J Am Heart Assoc 2020; 9:e014816. [PMID: 32172649 PMCID: PMC7335516 DOI: 10.1161/jaha.119.014816] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Dickkopf-1 and sclerostin have been implicated in atherosclerosis and vascular calcification. We aimed to quantify the association of their serum levels with incident cardiovascular disease (CVD) in the general population. Methods and Results Among 706 participants of the prospective, population-based Bruneck Study, mean±SD of serum levels were 44.5±14.7 pmol/L for dickkopf-1 and 47.1±17.5 pmol/L for sclerostin. The primary outcome was a composite CVD end point composed of ischemic or hemorrhagic stroke, transient ischemic attack, myocardial infarction, angina pectoris, peripheral vascular disease, and revascularization procedures. Over a median follow-up duration of 15.6 years, 179 CVD events occurred. For the primary CVD outcome, multivariable-adjusted hazard ratios (HRs) per SD higher level were 1.20 for dickkopf-1 (95% CI, 1.02-1.42; P=0.028) and 0.92 for sclerostin (95% CI, 0.78-1.08; P=0.286). Secondary outcome analyses revealed that the association of dickkopf-1 was primarily driven by ischemic and hemorrhagic stroke (67 events; HR, 1.37; 95% CI, 1.06-1.78; P=0.017), whereas no increase in risk was observed for transient ischemic attack (22 events; HR, 0.87; 95% CI, 0.53-1.44; P=0.593), myocardial infarction (45 events; HR, 1.10; 95% CI, 0.78-1.54; P=0.598), or for other CVD (45 events; HR, 1.25; 95% CI, 0.88-1.76; P=0.209). Conclusions In this prospective, population-based study, elevated baseline levels of dickkopf-1, but not sclerostin, were independently associated with incident cardiovascular events, which was mainly driven by stroke. Our findings support the hypothesis of a role of dickkopf-1 in the pathogenesis of CVD.
Collapse
Affiliation(s)
| | - Lena Tschiderer
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | | | | | - Stefan Brugger
- Departments of Internal Medicine Hospital of Bruneck Italy
| | - Peter Santer
- Departments of Laboratory Medicine Hospital of Bruneck Italy
| | - Johann Willeit
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Stefan Kiechl
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Peter Willeit
- Department of Neurology Medical University of Innsbruck Innsbruck Austria.,Department of Public Health and Primary Care University of Cambridge United Kingdom
| |
Collapse
|
10
|
Kuipers AL, Miljkovic I, Barinas‐Mitchell E, Nestlerode CS, Cvejkus RK, Wheeler VW, Zhang Y, Zmuda JM. Wnt Pathway Gene Expression Is Associated With Arterial Stiffness. J Am Heart Assoc 2020; 9:e014170. [PMID: 32013702 PMCID: PMC7033870 DOI: 10.1161/jaha.119.014170] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022]
Abstract
Background Animal and in vitro experiments implicate the Wnt pathway in cardiac development, fibrosis, vascular calcification, and atherosclerosis, but research in humans is lacking. We examined peripheral blood Wnt pathway gene expression and arterial stiffness in 369 healthy African ancestry men (mean age, 64 years). Methods and Results Gene expression was assessed using a custom Nanostring nCounter gene expression panel (N=43 genes) and normalized to housekeeping genes and background signal. Arterial stiffness was assessed via brachial-ankle pulse-wave velocity. Fourteen Wnt genes showed detectable expression and were tested individually as predictors of pulse-wave velocity using linear regression, adjusting for age, height, weight, blood pressure, medication use, resting heart rate, current smoking, alcohol intake, and sedentary lifestyle. Adenomatous polyposis coli regulator of Wnt signaling pathway (APC), glycogen synthase kinase 3β (GSK3B), and transcription factor 4 (TCF4) were significantly associated with arterial stiffness (P<0.05 for all). When entered into a single model, APC and TCF4 expression remained independently associated with arterial stiffness (P=0.04 and 0.003, respectively), and each explained ≈3% of the variance in pulse-wave velocity. Conclusions The current study establishes a novel association between in vivo expression of the Wnt pathway genes, APC and TCF4, with arterial stiffness in African ancestry men, a population at high risk of hypertensive vascular disease.
Collapse
Affiliation(s)
| | - Iva Miljkovic
- Department of EpidemiologyUniversity of PittsburghPittsburghPA
| | | | | | - Ryan K. Cvejkus
- Department of EpidemiologyUniversity of PittsburghPittsburghPA
| | | | - Yingze Zhang
- Department of MedicineUniversity of PittsburghPittsburghPA
| | - Joseph M. Zmuda
- Department of EpidemiologyUniversity of PittsburghPittsburghPA
| |
Collapse
|
11
|
Zhang CJ, Zhu N, Liu Z, Shi Z, Long J, Zu XY, Tang ZW, Hu ZY, Liao DF, Qin L. Wnt5a/Ror2 pathway contributes to the regulation of cholesterol homeostasis and inflammatory response in atherosclerosis. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158547. [PMID: 31678514 DOI: 10.1016/j.bbalip.2019.158547] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 09/24/2019] [Accepted: 10/15/2019] [Indexed: 01/26/2023]
Abstract
Atherosclerosis (AS) is characterized by lipids metabolism disorder and inflammatory response. Accumulating evidence has demonstrated that Wingless type 5a (Wnt5a) is implicated in cardiovascular diseases through non-canonical Wnt cascades. However, its precise role during the pathogenesis of AS is still unclear. Therefore, the present study aims to investigate the role and the underlying mechanism of Wnt5a/receptor tyrosine kinase-like orphan receptor 2 (Ror2) pathways in the promotion of AS process through affecting lipid accumulation and inflammation. In atherosclerotic clinical samples, Wnt5a levels were measured by using enzyme-linked immunosorbent assay (ELISA) assay. In vivo experiments were conducted by using apolipoprotein E knockout (apoE-/-) mice model. Vascular smooth muscle cells (VSMCs) were applied for in vitro studies. Wnt5a was highly expressed in both of atherosclerotic clinical samples and apoE-/- mice. The knockdown of Wnt5a significantly inhibited cholesterol accumulation and inflammatory response. Additionally, the lipopolysaccharide (LPS)-induced inflammation aggravated the cholesterol accumulation and decreased adenosine triphosphate (ATP)-binding cassette transporter A1 (ABCA1) expression in VSMCs. Depletion of intracellular cholesterol by β-cyclodextrin (β-CD) led to the upregulation of ABCA1 and the inhibition of inflammation. Conversely, the overexpression of Wnt5a inhibited ABCA1 expression, facilitated cholesterol accumulation, impared cholesterol efflux, promoted NF-κB nuclear translocation and the inflammatory cytokines secretion. Moreover, the knockdown of Ror2 increased ABCA1 expression and reduced Wnt5a-induced cholesterol accumulation and inflammatory responses. Furthermore, the knockdown of ABCA1 enhanced cholesterol accumulation and inflammatory response. Therefore, Wnt5a/Ror2 pathway was critical in regulating cholesterol homeostasis and inflammatory response, which might be a promising therapeutic target for AS therapy.
Collapse
Affiliation(s)
- Chan-Juan Zhang
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Neng Zhu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zheng Liu
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhe Shi
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jia Long
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xu-Yu Zu
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, China
| | - Zhen-Wang Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of University of South China, Heng Yang, Hunan, China
| | - Zhe-Yu Hu
- Department of Breast Medical Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Duan-Fang Liao
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Li Qin
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
12
|
Abstract
Clinical and preclinical studies over the past 3 decades have uncovered a multitude of signaling pathways involved in the initiation and progression of atherosclerosis. From these studies, signaling by proteins of the Wnt family has recently emerged as an important player in the development of atherosclerosis. Wnt signaling is characterized by a large number of ligands, receptors, and coreceptors and can be regulated at many different levels. Among Wnt modulators, the evolutionary conserved Dkk (Dickkopf) proteins, and especially Dkk-1, the founding member of the family, are the best characterized. The role of Dkks in the pathophysiology of the arterial wall is only partially understood, but their involvement in atherosclerosis is becoming increasingly evident. This review introduces recent key findings on Dkk proteins and their functions in atherosclerosis and discusses the potential importance of modulating Dkk signaling as part of a novel, improved strategy for preventing and treating atherosclerosis-related diseases.
Visual Overview—
An online visual overview is available for this article.
Collapse
Affiliation(s)
- Roberta Baetta
- From the Centro Cardiologico Monzino, IRCCS, Milano, Italy
| | - Cristina Banfi
- From the Centro Cardiologico Monzino, IRCCS, Milano, Italy
| |
Collapse
|
13
|
García-Gómez MC, Vilahur G. Osteoporosis and vascular calcification: A shared scenario. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2019; 32:33-42. [PMID: 31221532 DOI: 10.1016/j.arteri.2019.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 03/11/2019] [Indexed: 12/12/2022]
Abstract
Osteoporosis is a systemic skeletal disease, characterised by low bone mass and deterioration in the micro-architecture of bone tissue, which causes increased bone fragility and consequently greater susceptibility to fractures. It is the most frequent metabolic bone disease in our population, and fractures resulting from osteoporosis are becoming more common. Furthermore, vascular calcification is a recognised risk factor of cardiovascular morbidity and mortality that historically has been considered a passive and degenerative process. However, it is currently recognised as an active process, which has histopathological characteristics, mineral composition and initiation and development mechanisms characteristic of bone formation. Paradoxically, patients with osteoporosis frequently show vascular calcifications. Traditionally, they have been considered as independent processes related to age, although more recent epidemiological studies have shown that there is a close relationship between the loss of bone mass and vascular calcification, regardless of age. In fact, both conditions share risk factors and pathophysiological mechanisms. These include the relationship between proteins of bone origin, such as osteopontin and osteoprotegerin (OPG), with vascular pathology, and the intercellular protein system RANK/RANKL/OPG and the Wnt signalling pathway. The mechanisms linked in both pathologies should be considered in clinical decisions, given that treatments for osteoporosis could have unforeseen effects on vascular calcification, and vice versa. In short, a better understanding of the relationship between both entities can help in proposing strategies to reduce the increasing prevalence of vascular calcification and osteoporosis in the aging population.
Collapse
Affiliation(s)
| | - Gemma Vilahur
- Programa ICCC-Institut de Recerca Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, España; CIBERCV Instituto de Salud Carlos III, Madrid, España.
| |
Collapse
|
14
|
Vallée A, Vallée JN, Lecarpentier Y. Metabolic reprogramming in atherosclerosis: Opposed interplay between the canonical WNT/β-catenin pathway and PPARγ. J Mol Cell Cardiol 2019; 133:36-46. [PMID: 31153873 DOI: 10.1016/j.yjmcc.2019.05.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 01/08/2023]
Abstract
Atherosclerosis, a chronic inflammatory and age-related disease, is a complex mechanism presenting a dysregulation of vessel structures. During this process, the canonical WNT/β-catenin pathway is increased whereas PPARγ is downregulated. The two systems act in an opposite manner. This paper reviews the opposing interplay of these systems and their metabolic-reprogramming pathway in atherosclerosis. Activation of the WNT/β-catenin pathway enhances the transcription of targets involved in inflammation, endothelial dysfunction, the proliferation of vascular smooth muscle cells, and vascular calcification. This complex mechanism, which is partly controlled by the WNT/β-catenin pathway, presents several metabolic dysfunctions. This phenomenon, called aerobic glycolysis (or the Warburg effect), consists of a shift in ATP production from mitochondrial oxidative phosphorylation to aerobic glycolysis, leading to the overproduction of intracellular lactate. This mechanism is partially due to the injury of mitochondrial respiration and an increase in the glycolytic pathway. In contrast, PPARγ agonists downregulate the WNT/β-catenin pathway. Therefore, the development of therapeutic targets, such as PPARγ agonists, for the treatment of atherosclerosis could be an interesting and innovative way of counteracting the canonical WNT pathway.
Collapse
Affiliation(s)
- Alexandre Vallée
- Diagnosis and Therapeutic Center, Hypertension and Cardiovascular Prevention Unit, Hotel-Dieu Hospital, AP-HP, Université Paris Descartes, Paris, France.
| | - Jean-Noël Vallée
- Centre Hospitalier Universitaire (CHU) Amiens Picardie, Université Picardie Jules Verne (UPJV), 80054 Amiens, France; Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, Poitiers, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), 6-8 rue Saint-fiacre, 77100 Meaux, France
| |
Collapse
|
15
|
Li H, Zhao Q, Chang L, Wei C, Bei H, Yin Y, Chen M, Wang H, Liang J, Wu Y. LncRNA MALAT1 modulates ox-LDL induced EndMT through the Wnt/β-catenin signaling pathway. Lipids Health Dis 2019; 18:62. [PMID: 30871555 PMCID: PMC6417088 DOI: 10.1186/s12944-019-1006-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/06/2019] [Indexed: 02/05/2023] Open
Abstract
Background Endothelial-to-mesenchymal transition (EndMT) plays significant roles in atherosclerosis, but the regulatory mechanisms involving lncRNAs remain to be elucidated. Here we sort to identify the role of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in ox-LDL-induced EndMT. Methods The atherosclerosis model was established by feeding ApoE−/− mice with high-fat diet, and the levels of lncRNA MALAT1 in mouse arterial tissue were detected by RT-qPCR. Cell model was established by treating human umbilical vein endothelial cells (HUVECs) with ox-LDL, and the levels of EndMT markers, such as CD31, vWF, α-SMA and Vimentin and lncRNA MALAT1 levels were detected and their correlations were analyzed. The role of MALAT1 in EndMT and its dependence on Wnt/β-catenin signaling pathway was further detected by knocking down or overexpressing MALAT1. Results MALAT1 was upregulated in high-fat food fed ApoE−/− mice. HUVECs treated with ox-LDL showed a significant decrease in expression of CD31 and vWF, a significant increase in expression of α-SMA and vimentin, and upregulated MALAT1. An increased MALAT1 level facilitated the nuclear translocation of β-catenin induced by ox-LDL. Inhibition of MALAT1 expression reversed nuclear translocation of β-catenin and EndMT. Moreover, overexpression of MALAT1 enhanced the effects of ox-LDL on HUVEC EndMT and Wnt/β-catenin signaling activation. Conclusions Our study revealed that the pathological EndMT required the activation of the MALAT1-dependent Wnt/β-catenin signaling pathway, which may be important for the onset of atherosclerosis. Trial registration Not applicable.
Collapse
Affiliation(s)
- Hongrong Li
- Hebei Medical University, No. 361, Zhongshan East Road, Changan District, Shijiazhuang, 050017, China
| | - Qifei Zhao
- Hebei Medical University, No. 361, Zhongshan East Road, Changan District, Shijiazhuang, 050017, China
| | - Liping Chang
- National Key Laboratory of Luobing Research and Innovative Chinese Medicine, Shijiazhuang, 050035, China
| | - Cong Wei
- Hebei Medical University, No. 361, Zhongshan East Road, Changan District, Shijiazhuang, 050017, China.,Hebei Key Laboratory of Luobing, Shijiazhuang, 050035, China
| | - Hongying Bei
- Yiling Hospital of Hebei Medical University, The Key Laboratory of State Administration of Traditional Chinese Medicine, Shijiazhuang, 050091, China
| | - Yujie Yin
- Yiling Hospital of Hebei Medical University, The Key Laboratory of State Administration of Traditional Chinese Medicine, Shijiazhuang, 050091, China.,Hebei University of Chinese Medicine, Shijiazhuang, 050090, China
| | - Meng Chen
- National Key Laboratory of Luobing Research and Innovative Chinese Medicine, Shijiazhuang, 050035, China
| | - Hongtao Wang
- National Key Laboratory of Luobing Research and Innovative Chinese Medicine, Shijiazhuang, 050035, China
| | - Junqing Liang
- National Key Laboratory of Luobing Research and Innovative Chinese Medicine, Shijiazhuang, 050035, China
| | - Yiling Wu
- Hebei Medical University, No. 361, Zhongshan East Road, Changan District, Shijiazhuang, 050017, China. .,Yiling Hospital of Hebei Medical University, The Key Laboratory of State Administration of Traditional Chinese Medicine, Shijiazhuang, 050091, China.
| |
Collapse
|
16
|
Bartoli-Leonard F, Wilkinson FL, Langford-Smith AWW, Alexander MY, Weston R. The Interplay of SIRT1 and Wnt Signaling in Vascular Calcification. Front Cardiovasc Med 2018; 5:183. [PMID: 30619890 PMCID: PMC6305318 DOI: 10.3389/fcvm.2018.00183] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/04/2018] [Indexed: 12/24/2022] Open
Abstract
Vascular calcification is a major health risk and is highly correlated with atherosclerosis, diabetes, and chronic kidney disease. The development of vascular calcification is an active and complex process linked with a multitude of signaling pathways, which regulate promoters and inhibitors of osteogenesis, the balance of which become deregulated in disease conditions. SIRT1, a protein deacetylase, known to be protective in inhibiting oxidative stress and inflammation within the vessel wall, has been shown as a possible key player in modulating the cell-fate determining canonical Wnt signaling pathways. Suppression of SIRT1 has been reported in patients suffering with cardiovascular pathologies, suggesting that the sustained acetylation of osteogenic factors could contribute to their activation and in turn, lead to the progression of calcification. There is clear evidence of the synergy between β-Catenin and elevated Runx2, and with Wnt signaling being β-Catenin dependent, further understanding is needed as to how these molecular pathways converge and interact, in order to provide novel insight into the mechanism by which smooth muscle cells switch to an osteogenic differentiation programme. Therefore, this review will describe the current concepts of pathological soft tissue mineralization, with a focus on the contribution of SIRT1 as a regulator of Wnt signaling and its targets, discussing SIRT1 as a potential target for manipulation and therapy.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Fiona L Wilkinson
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Alex W W Langford-Smith
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - M Y Alexander
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| | - Ria Weston
- Translational Cardiovascular Science, Centre for Bioscience, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
17
|
Appelman-Dijkstra NM, Papapoulos SE. Clinical advantages and disadvantages of anabolic bone therapies targeting the WNT pathway. Nat Rev Endocrinol 2018; 14:605-623. [PMID: 30181608 DOI: 10.1038/s41574-018-0087-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The WNT signalling pathway is a key regulator of bone metabolism, particularly bone formation, which has helped to define the role of osteocytes - the most abundant bone cells - as orchestrators of bone remodelling. Several molecules involved in the control of the WNT signalling pathway have been identified as potential targets for the development of bone-building therapeutics for patients with osteoporosis. Several of these molecules have been investigated in animal models, but only inhibitors of sclerostin (which is produced by osteocytes) have been investigated in phase III clinical studies. Here, we review the rationale for these developments and the specificity and potential off-target actions of WNT-based therapeutics. We also describe the available preclinical and clinical studies and discuss the benefits and risks of using sclerostin inhibitors for the management of patients with osteoporosis.
Collapse
|
18
|
Carbone F, Montecucco F. Novel cardiovascular risk biomarkers in carotid atherogenesis. Biomark Med 2018; 12:1065-1067. [PMID: 30227729 DOI: 10.2217/bmm-2018-0198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genova 10 Largo Benzi, 16132, Genoa, Italy & Center of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| |
Collapse
|
19
|
Zhang P, Hua L, Hou H, Du X, He Z, Liu M, Hu X, Yan N. Sphingomyelin synthase 2 promotes H2O2-induced endothelial dysfunction by activating the Wnt/β-catenin signaling pathway. Int J Mol Med 2018; 42:3344-3354. [PMID: 30272329 PMCID: PMC6202097 DOI: 10.3892/ijmm.2018.3888] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 09/17/2018] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis (AS) is the primary cause of various cardiovascular and cerebrovascular diseases and has high morbidity and mortality rates. Oxidative stress-induced endothelial cells (ECs) dysfunction is the pathological basis of AS. In addition, sphingomyelin (SM) and the Wnt/β-catenin signaling pathway are considered to be closely associated with AS; however, the specific mechanism is not clear. Therefore, the present study investigated whether SM may induce ECs dysfunction through the Wnt/β-catenin signaling pathway. Firstly, a sphingomyelin synthase 2 (SMS2) overexpression cell model was constructed. It was identified that the expression of SMS2 was increased when ECs were treated with H2O2. In addition, these results demonstrated that SMS2 overexpression promoted apoptosis and macrophage adhesion of H2O2-induced ECs, thereby increasing the expression of β-catenin. Furthermore, SMS activity was inhibited with Dy105, combined with simultaneous treatment with LiCl or H2O2. This additionally confirmed that Dy105 significantly inhibited SMS activity and decreased the level of ECs dysfunction and β-catenin content; however, LiCl served a key role in activating the Wnt/β-catenin signaling pathway to promote ECs dysfunction. Collectively, these results suggested that SMS2 overexpression may promote ECs dysfunction by activating the Wnt/β-catenin signaling pathway, while Dy105 may inhibit the evolution of oxidative stress-induced dysfunction.
Collapse
Affiliation(s)
- Panpan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lingyue Hua
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Huan Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xingyue Du
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhiqiang He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Menghan Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaojuan Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Nianlong Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
20
|
Wang F, Liu Z, Park SH, Gwag T, Lu W, Ma M, Sui Y, Zhou C. Myeloid β-Catenin Deficiency Exacerbates Atherosclerosis in Low-Density Lipoprotein Receptor-Deficient Mice. Arterioscler Thromb Vasc Biol 2018; 38:1468-1478. [PMID: 29724817 PMCID: PMC6023740 DOI: 10.1161/atvbaha.118.311059] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 04/18/2018] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— The Wnt/β-catenin signaling is an ancient and evolutionarily conserved pathway that regulates essential aspects of cell differentiation, proliferation, migration and polarity. Canonical Wnt/β-catenin signaling has also been implicated in the pathogenesis of atherosclerosis. Macrophage is one of the major cell types involved in the initiation and progression of atherosclerosis, but the role of macrophage β-catenin in atherosclerosis remains elusive. This study aims to investigate the impact of β-catenin expression on macrophage functions and atherosclerosis development. Approach and Results— To investigate the role of macrophage canonical Wnt/β-catenin signaling in atherogenesis, we generated β-cateninΔmyeLDLR−/− mice (low-density lipoprotein receptor–deficient mice with myeloid-specific β-catenin deficiency). As expected, deletion of β-catenin decreased macrophage adhesion and migration properties in vitro. However, deficiency of β-catenin significantly increased atherosclerotic lesion areas in the aortic root of LDLR−/− (low-density lipoprotein receptor–deficient) mice without affecting the plasma lipid levels and atherosclerotic plaque composition. Mechanistic studies revealed that β-catenin can regulate activation of STAT (signal transducer and activator of transcription) pathway in macrophages, and ablation of β-catenin resulted in STAT3 downregulation and STAT1 activation, leading to elevated macrophage inflammatory responses and increased atherosclerosis. Conclusions— This study demonstrates a critical role of myeloid β-catenin expression in atherosclerosis by modulating macrophage inflammatory responses.
Collapse
Affiliation(s)
- Fang Wang
- From the Department of Pharmacology and Nutritional Sciences (F.W., Z.L., S.-H.P., T.G., W.L., M.M., Y.S., C.Z.)
| | - Zun Liu
- From the Department of Pharmacology and Nutritional Sciences (F.W., Z.L., S.-H.P., T.G., W.L., M.M., Y.S., C.Z.)
| | - Se-Hyung Park
- From the Department of Pharmacology and Nutritional Sciences (F.W., Z.L., S.-H.P., T.G., W.L., M.M., Y.S., C.Z.)
| | - Taesik Gwag
- From the Department of Pharmacology and Nutritional Sciences (F.W., Z.L., S.-H.P., T.G., W.L., M.M., Y.S., C.Z.)
| | - Weiwei Lu
- From the Department of Pharmacology and Nutritional Sciences (F.W., Z.L., S.-H.P., T.G., W.L., M.M., Y.S., C.Z.)
| | - Murong Ma
- From the Department of Pharmacology and Nutritional Sciences (F.W., Z.L., S.-H.P., T.G., W.L., M.M., Y.S., C.Z.)
| | - Yipeng Sui
- From the Department of Pharmacology and Nutritional Sciences (F.W., Z.L., S.-H.P., T.G., W.L., M.M., Y.S., C.Z.)
| | - Changcheng Zhou
- From the Department of Pharmacology and Nutritional Sciences (F.W., Z.L., S.-H.P., T.G., W.L., M.M., Y.S., C.Z.)
- Saha Cardiovascular Research Center (C.Z.), University of Kentucky, Lexington
| |
Collapse
|