1
|
Somanader DVN, Zhao P, Widdop RE, Samuel CS. The involvement of the Wnt/β-catenin signaling cascade in fibrosis progression and its therapeutic targeting by relaxin. Biochem Pharmacol 2024; 223:116130. [PMID: 38490518 DOI: 10.1016/j.bcp.2024.116130] [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: 11/29/2023] [Revised: 02/06/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Organ scarring, referred to as fibrosis, results from a failed wound-healing response to chronic tissue injury and is characterised by the aberrant accumulation of various extracellular matrix (ECM) components. Once established, fibrosis is recognised as a hallmark of stiffened and dysfunctional tissues, hence, various fibrosis-related diseases collectively contribute to high morbidity and mortality in developed countries. Despite this, these diseases are ineffectively treated by currently-available medications. The pro-fibrotic cytokine, transforming growth factor (TGF)-β1, has emerged as the master regulator of fibrosis progression, owing to its ability to promote various factors and processes that facilitate rapid ECM synthesis and deposition, whilst negating ECM degradation. TGF-β1 signal transduction is tightly controlled by canonical (Smad-dependent) and non-canonical (MAP kinase- and Rho-associated protein kinase-dependent) intracellular protein activity, whereas its pro-fibrotic actions can also be facilitated by the Wnt/β-catenin pathway. This review outlines the pathological sequence of events and contributing roles of TGF-β1 in the progression of fibrosis, and how the Wnt/β-catenin pathway contributes to tissue repair in acute disease settings, but to fibrosis and related tissue dysfunction in synergy with TGF-β1 in chronic diseases. It also outlines the anti-fibrotic and related signal transduction mechanisms of the hormone, relaxin, that are mediated via its negative modulation of TGF-β1 and Wnt/β-catenin signaling, but through the promotion of Wnt/β-catenin activity in acute disease settings. Collectively, this highlights that the crosstalk between TGF-β1 signal transduction and the Wnt/β-catenin cascade may provide a therapeutic target that can be exploited to broadly treat and reverse established fibrosis.
Collapse
Affiliation(s)
- Deidree V N Somanader
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Peishen Zhao
- Drug Discovery Biology Program, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3052, Australia.
| |
Collapse
|
2
|
Xu X, Zhang B, Wang Y, Shi S, Lv J, Fu Z, Gao X, Li Y, Wu H, Song Q. Renal fibrosis in type 2 cardiorenal syndrome: An update on mechanisms and therapeutic opportunities. Biomed Pharmacother 2023; 164:114901. [PMID: 37224755 DOI: 10.1016/j.biopha.2023.114901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/26/2023] Open
Abstract
Cardiorenal syndrome (CRS) is a state of coexisting heart failure and renal insufficiency in which acute or chronic dysfunction of the heart or kidney lead to acute or chronic dysfunction of the other organ.It was found that renal fibrosis is an important pathological process in the progression of type 2 CRS to end-stage renal disease, and progressive renal impairment accelerates the deterioration of cardiac function and significantly increases the hospitalization and mortality rates of patients. Previous studies have found that Hemodynamic Aiteration, RAAS Overactivation, SNS Dysfunction, Endothelial Dysfunction and Imbalance of natriuretic peptide system contribute to the development of renal disease in the decompensated phase of heart failure, but the exact mechanisms is not clear. Therefore, in this review, we focus on the molecular pathways involved in the development of renal fibrosis due to heart failure and identify the canonical and non-canonical TGF-β signaling pathways and hypoxia-sensing pathways, oxidative stress, endoplasmic reticulum stress, pro-inflammatory cytokines and chemokines as important triggers and regulators of fibrosis development, and summarize the therapeutic approaches for the above signaling pathways, including SB-525334 Sfrp1, DKK1, IMC, rosarostat, 4-PBA, etc. In addition, some potential natural drugs for this disease are also summarized, including SQD4S2, Wogonin, Astragaloside, etc.
Collapse
Affiliation(s)
- Xia Xu
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingxuan Zhang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yajiao Wang
- College of Traditional Chinese Medicine, China Academy of Chinese Medical Science, Beijing, China
| | - Shuqing Shi
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiayu Lv
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyue Fu
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Xiya Gao
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Yumeng Li
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Huaqin Wu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qingqiao Song
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
3
|
Jiang B, Zhou X, Yang T, Wang L, Feng L, Wang Z, Xu J, Jing W, Wang T, Su H, Yang G, Zhang Z. The role of autophagy in cardiovascular disease: Cross-interference of signaling pathways and underlying therapeutic targets. Front Cardiovasc Med 2023; 10:1088575. [PMID: 37063954 PMCID: PMC10090687 DOI: 10.3389/fcvm.2023.1088575] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
Autophagy is a conserved lysosomal pathway for the degradation of cytoplasmic proteins and organelles, which realizes the metabolic needs of cells and the renewal of organelles. Autophagy-related genes (ATGs) are the main molecular mechanisms controlling autophagy, and their functions can coordinate the whole autophagic process. Autophagy can also play a role in cardiovascular disease through several key signaling pathways, including PI3K/Akt/mTOR, IGF/EGF, AMPK/mTOR, MAPKs, p53, Nrf2/p62, Wnt/β-catenin and NF-κB pathways. In this paper, we reviewed the signaling pathway of cross-interference between autophagy and cardiovascular diseases, and analyzed the development status of novel cardiovascular disease treatment by targeting the core molecular mechanism of autophagy as well as the critical signaling pathway. Induction or inhibition of autophagy through molecular mechanisms and signaling pathways can provide therapeutic benefits for patients. Meanwhile, we hope to provide a unique insight into cardiovascular treatment strategies by understanding the molecular mechanism and signaling pathway of crosstalk between autophagy and cardiovascular diseases.
Collapse
Affiliation(s)
- Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xuan Zhou
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Yang
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linlin Wang
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Longfei Feng
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zheng Wang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jin Xu
- Department of First Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Wang
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Haixiang Su
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - GuoWei Yang
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| | - Zheng Zhang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
4
|
Zhang Y, Fu C, Zhao S, Jiang H, Li W, Liu X. PRELP promotes myocardial fibrosis and ventricular remodelling after acute myocardial infarction by the wnt/β-catenin signalling pathway. Cardiovasc J Afr 2022; 33:228-233. [PMID: 35788244 PMCID: PMC9887443 DOI: 10.5830/cvja-2022-001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 12/20/2021] [Indexed: 10/08/2023] Open
Abstract
OBJECTIVES Proline/arginine-rich end leucine-rich repeat protein (PRELP) has been reported to contribute to the remodelling of cardiovascular tissues in the ischaemia-reperfusion injury model. However, research is lacking on the role of PRELP in myocardial fibrosis and ventricular remodelling, and the mechanism through which PRELP brings about these changes is not clear. This study aimed to evaluate the role of PRELP in ventricular remodelling and myocardial fibrosis following acute myocardial infarction (AMI) and to explore the underlying mechanism. METHODS In this study, we established AMI mouse and cellculture models in an oxygen-glucose deprivation environment. RESULTS We found that over-expression of PRELP increased the infarct size and interstitial fibrotic area. Expression of the wnt/β-catenin pathway molecules, which are downstream of PRELP, increased more in the PRELP over-expression group than in the AMI group. CONCLUSIONS Our results showed that PRELP, through the wnt/β-catenin signalling pathway, led to myocardial fibrosis and ventricular remodelling following AMI.
Collapse
Affiliation(s)
- Yu Zhang
- Shandong Key Laboratory of Cardiovascular Proteomics, Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.
| | - Chunli Fu
- Shandong Key Laboratory of Cardiovascular Proteomics, Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Shaohua Zhao
- Shandong Key Laboratory of Cardiovascular Proteomics, Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Honglei Jiang
- Department of Cardiology, Shandong Provincial Western Hospital, Jinan, Shandong, People's Republic of China
| | - Wei Li
- Department of Anesthesia, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Xiangju Liu
- Shandong Key Laboratory of Cardiovascular Proteomics, Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
5
|
Feng D, Li J, Guo L, Liu J, Wang S, Ma X, Song Y, Liu J, Hao E. DDX3X alleviates doxorubicin-induced cardiotoxicity by regulating Wnt/β-catenin signaling pathway in an in vitro model. J Biochem Mol Toxicol 2022; 36:e23077. [PMID: 35467791 PMCID: PMC9539463 DOI: 10.1002/jbt.23077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/21/2022] [Accepted: 04/01/2022] [Indexed: 11/05/2022]
Abstract
The life-threatening adverse effects of doxorubicin (Dox) caused by its cardiotoxic properties limit its clinical application. DDX3X has been shown to participate in a variety of physiological processes, and it acts as a regulator of Wnt/β-catenin signaling. However, the role of DDX3X in Dox-induced cardiotoxicity (DIC) remains unclear. In this study, we found that DDX3X expression was significantly decreased in H9c2 cardiomyocytes treated with Dox. Ddx3x knockdown and RK-33 (DDX3X ATPase activity inhibitor) pretreatment exacerbated cardiomyocyte apoptosis and mitochondrial dysfunction induced by Dox treatment. In contrast, Ddx3x overexpression ameliorated the DIC response. Moreover, Wnt/β-catenin signaling in cardiomyocytes treated with Dox was suppressed, but this suppression was reversed by Ddx3x overexpression. Overall, this study demonstrated that DDX3X plays a protective role in DIC by activating Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Dandan Feng
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Jiang Li
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Liang Guo
- Department of AnesthesiologyThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
| | - Jing Liu
- Laboratory of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan HospitalThe First Affiliated Hospital of Shandong First Medical UniversityJinanChina
| | - Shaochen Wang
- Department of Cardiology, Shandong Provincial Qianfoshan HospitalShandong First Medical UniversityJinanChina
| | - Xiuyuan Ma
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Yunxuan Song
- Department of Cardiology, Shandong Provincial Qianfoshan HospitalShandong First Medical UniversityJinanChina
| | - Ju Liu
- Laboratory of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan HospitalThe First Affiliated Hospital of Shandong First Medical UniversityJinanChina
| | - Enkui Hao
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| |
Collapse
|
6
|
Liu J, Zheng X, Zhang C, Zhang C, Bu P. Lcz696 Alleviates Myocardial Fibrosis After Myocardial Infarction Through the sFRP-1/Wnt/β-Catenin Signaling Pathway. Front Pharmacol 2021; 12:724147. [PMID: 34539406 PMCID: PMC8443774 DOI: 10.3389/fphar.2021.724147] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Lcz696 (ARNI, angiotensin receptor–neprilysin inhibitor; sacubitril/valsartan) shows an inhibitory effect on fibrosis after myocardial infarction (MI). However, the underlying signaling mechanisms are poorly understood. The Wnt/β-catenin signaling pathway is activated after MI and participates in the process of myocardial fibrosis. Here, we aimed to assess the efficacy of ARNI for alleviating myocardial fibrosis after MI and hypothesized that ARNI alleviates myocardial fibrosis by inhibiting the Wnt/β-catenin signaling pathway and overexpressing sFRP-1, an inhibitor of the Wnt/β-catenin signaling pathway. Methods: Mice randomized at 1 week post-MI were administered lcz696 (60 mg/kg, n = 21), valsartan (30 mg/kg, n = 19), or corn oil (n = 13) orally for 4 weeks, while the sham-operated group received vehicle (corn oil, n = 19). Cardiac function and extent of myocardial fibrosis were measured. Western blotting and quantitative real-time polymerase chain reaction were used to detect the expression of Wnt/β-catenin pathway-related proteins. Furthermore, primary myocardial fibroblasts were stimulated with angiotensin II (Ang II) and cultured with lcz696 and the sFRP-1 inhibitor way316606 to detect the expression of Wnt/β-catenin pathway proteins. Results: Both lcz696 and valsartan alleviated myocardial fibrosis and improved cardiac function, but lcz696 had superior efficiency compared to valsartan. Furthermore, β-catenin expression was inhibited and sFRP-1 was overexpressed after drug treatment, which could be significantly improved by lcz696 in mice. In addition, lcz696 inhibited β-catenin expression in AngII-stimulated myocardial fibroblasts, and β-catenin expression increased after the inhibition of sFRP-1. Conclusion: ARNI alleviated cardiac fibrosis and cardiac remodeling by inhibiting the Wnt/β-catenin signaling pathway. In addition, ARNI can lead to overexpression of sFRP-1, which is an inhibitor of the Wnt/β-catenin signaling pathway. These results indicate a new therapeutic target of ARNI to improve myocardial fibrosis and prevent myocardial remodeling.
Collapse
Affiliation(s)
- Jing Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Cardiology, Heze Municipal Hospital, Heze, China
| | - Xuehui Zheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chen Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunmei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peili Bu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
7
|
Sfrp1 protects against acute myocardial ischemia (AMI) injury in aged mice by inhibiting the Wnt/β-catenin signaling pathway. J Cardiothorac Surg 2021; 16:12. [PMID: 33468190 PMCID: PMC7814560 DOI: 10.1186/s13019-020-01389-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/28/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aged patients suffering from acute myocardial ischemia (AMI) exhibit an increased mortality rate and worse prognosis, and a more effective treatment is currently in need. In the present study, we investigated potent targets related to Wnt/β-catenin pathway deregulation for AMI injury treatment. METHODS In the present study, AAV-Sfrp1 was transduced into the myocardium of aged mice, and an AMI model was established in these aged mice to study the effect and molecular mechanism of Sfrp1 overexpression on AMI-induced injury. RESULTS The results showed that Sfrp1 was successfully overexpressed in the myocardium of aged mice and remarkably reduced Wnt/β-catenin pathway activity in aged mice after AMI, effectively reducing the degree of myocardial fibrosis, inhibiting cardiomyocyte apoptosis, and improving cardiac function. We revealed that the exogenous introduction of Sfrp1 could be considered a promising strategy for improving post-AMI injury in aged mice by inhibiting Wnt/β-catenin pathway activity. CONCLUSIONS In conclusion, the Wnt/β-catenin pathway potentially represents a key target in AMI in aged mice. Sfrp1 might be used as a small molecule gene therapy drug to improve heart function, reduce the degree of myocardial fibrosis, inhibit cardiomyocyte apoptosis and reduce AMI injury in aged mice by inhibiting the Wnt/β-catenin pathway, thereby effectively protecting aged hearts from AMI injury.
Collapse
|
8
|
The effect of nutraceuticals on multiple signaling pathways in cardiac fibrosis injury and repair. Heart Fail Rev 2020; 27:321-336. [PMID: 32495263 DOI: 10.1007/s10741-020-09980-6] [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] [Indexed: 10/24/2022]
Abstract
Cardiac fibrosis is one of the most common pathological conditions caused by different heart diseases, including myocardial infarction and diabetic cardiomyopathy. Cardiovascular disease is one of the major causes of mortality worldwide. Cardiac fibrosis is caused by different processes, including inflammatory reactions and oxidative stress. The process of fibrosis begins by changing the balance between production and destruction of extracellular matrix components and stimulating the proliferation and differentiation of cardiac fibroblasts. Many studies have focused on finding drugs with less adverse effects for the treatment of cardiovascular disease. Some studies show that nutraceuticals are effective in preventing and treating diseases, including cardiovascular disease, and that they can reduce the risk. However, big clinical studies to prove the therapeutic properties of all these substances and their adverse effects are lacking so far. Therefore, in this review, we tried to summarize the knowledge on pathways and mechanisms of several nutraceuticals which have shown their usefulness in the prevention of cardiac fibrosis.
Collapse
|
9
|
Eid RA, Khalil MA, Alkhateeb MA, Eleawa SM, Zaki MSA, El-Kott AF, Al-Shraim M, El-Sayed F, Eldeen MA, Bin-Meferij MM, Awaji KME, Shatoor AS. Exendin-4 Attenuates Remodeling in the Remote Myocardium of Rats After an Acute Myocardial Infarction by Activating β-Arrestin-2, Protein Phosphatase 2A, and Glycogen Synthase Kinase-3 and Inhibiting β-Catenin. Cardiovasc Drugs Ther 2020; 35:1095-1110. [PMID: 32474680 DOI: 10.1007/s10557-020-07006-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE This study tested if the protective anti-remodeling effect of GLP-1 agonist Exendin-4 after an acute myocardial infarction (MI) in rats involves inhibition of the Wnt1/β-catenin signaling pathway. METHODS Rats were divided into sham, sham + Exendin-4 (10 μg/day, i.p), MI, and MI + Exendin-4. MI was introduced to rats by permanent left anterior descending coronary artery (LAD) ligation. RESULTS On day 7 post-infraction, MI rats showed LV dysfunction with higher serum levels of cardiac markers. Their remote myocardia showed increased mRNA and protein levels of collagen I/III with higher levels of reactive oxygen species (ROS) and inflammatory cytokines, as well as protein levels of Wnt1, phospho-Akt, transforming growth factor (TGF-β1), Smad, phospho-Smad3, α-SMA, caspase-3, and Bax. They also showed higher protein levels of phospho-glycogen synthase kinase-3β (p-GSK3β), as well as total, phosphorylated, and nuclear β-catenin with a concomitant decrease in the levels of cyclic adenosine monophosphate (cAMP), mRNA of manganese superoxide dismutase (MnSOD), and protein levels of Bcl-2, β-arrestin-2, and protein phosphatase-2 (PP2A). Administration of Exendin-4 to MI rats reduced the infarct size and reversed the aforementioned signaling molecules without altering protein levels of TGF-1β and Wnt1 or Akt activation. Interestingly, Exendin-4 increased mRNA levels of MnSOD, protein levels of β-arrestin-2 and PP2A, and β-catenin phosphorylation but reduced the phosphorylation of GSK3β and Smad3, and total β-catenin levels in the LV of control rats. CONCLUSION Exendin-4 inhibits the remodeling in the remote myocardium of rats following acute MI by attenuating β-catenin activation and activating β-arrestin-2, PP2A, and GSK3β. Graphical Abstract A graphical abstract that illustrates the mechanisms by which Exendin-4 inhibits cardiac remodeling in remote myocardium of left ventricle MI-induced rats. Mechanisms are assumed to occur in the cardiomyocytes and/or other resident cells such as fibroblast. Β-catenin activation and nuclear translocation are associated with increased synthesis of inflammatory cytokines and transforming growth factor β-1 (TGF-β1). GSK3β is inhibited by phosphorylation at Ser9. Under normal conditions, β-catenin is degraded in the cytoplasm by the active GSK3β-dependent degradation complex (un-phosphorylated) which usually phosphorylates β-catenin at Ser33/37/Thr41. After MI, TGF-β1, and Wnt 1 levels are significantly increased, the overproduction of Wnt1 induces β-catenin stabilization and nuclear translocation through increasing the phosphorylation of disheveled (DVL) protein which in turn phosphorylates and inhibits GSK3β. TGF-β1 stimulates the phosphorylation of Smad-3 and subsequent nuclear translocation to activate the transcription of collage 1/III and α-smooth muscle actin (α-SMA). Besides, TGF-β1 stabilizes cytoplasmic β-catenin levels indirectly by phosphorylation of Akt at Thr308-induced inhibition of GSK3β by increasing phosphorylation of Ser9. Exendin-4, and possibly through G protein-coupled receptors (GPCRs), increases levels of cAMP and upregulates β-arrestin-2 levels. Both can result in a positive inotropic effect. Besides, β-arrestin-2 can stimulate PP2A to dephosphorylation Smad3 (inhibition) and GSK3β (activation), thus reduces fibrosis and prevents the activation of β-catenin and collagen deposition.
Collapse
Affiliation(s)
- Refaat A Eid
- Department of Pathology, College of Medicine, King Khalid University, P.O. 641, Abha, 61421, Saudi Arabia.
| | - Mohammad Adnan Khalil
- Department of Basic Medical Sciences, Faculty of Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Mahmoud A Alkhateeb
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Samy M Eleawa
- Department of Applied Medical Sciences, College of Health Sciences, PAAET, Shuwaikh, Kuwait
| | - Mohamed Samir Ahmed Zaki
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia.,Department of Histology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Attalla Farag El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.,Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Mubarak Al-Shraim
- Department of Pathology, College of Medicine, King Khalid University, P.O. 641, Abha, 61421, Saudi Arabia
| | - Fahmy El-Sayed
- Department of Pathology, College of Medicine, King Khalid University, P.O. 641, Abha, 61421, Saudi Arabia
| | - Muhammad Alaa Eldeen
- Department of Biology, Physiology Section, Faculty of Science, Zagazig University, Zagazig, Egypt
| | | | - Khalid M E Awaji
- Clinical laboratories Department, Asser Central Hospital, Abha, Saudi Arabia
| | - Abdullah S Shatoor
- Department of Clinical Cardiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
10
|
Badimon L, Casaní L, Camino-Lopez S, Juan-Babot O, Borrell-Pages M. GSK3β inhibition and canonical Wnt signaling in mice hearts after myocardial ischemic damage. PLoS One 2019; 14:e0218098. [PMID: 31220102 PMCID: PMC6586285 DOI: 10.1371/journal.pone.0218098] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 05/25/2019] [Indexed: 12/22/2022] Open
Abstract
Aims Myocardial infarction induces myocardial injury and tissue damage. During myocardial infarction strong cellular response is initiated to salvage the damaged tissues. This response is associated with the induction of different signaling pathways. Of these, the canonical Wnt signaling is increasingly important for its prosurvival cellular role, making it a good candidate for the search of new molecular targets to develop therapies to prevent heart failure in infarcted patients. Methods Herein we report that GSK3β regulates the canonical Wnt signaling in C57Bl6 mice hearts. GSK3β is a canonical Wnt pathway inhibitor. Using GSK3β inhibitors and inducing myocardial injury (MI) in Lrp5-/- mice model we show that GSK3β phosphorylation levels regulate downstream canonical Wnt pathway genes in the ischemic heart. In the setting of MI, myocardial damage assessment usually correlates with functional and clinical outcomes. Therefore, we measured myocardial injury size in Wt and Lrp5-/- mice in the presence and absence of two different GSK3 inhibitors prior to MI. Myocardial injury was independent of GSK3 inhibitor treatments and GSK3β expression levels. Results These studies support a central role for GSK3β in the activation of the canonical Wnt pathway in the Wt heart. Although LRP5 is protective against myocardial injury, GSK3β expression levels do not regulate heart damage.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Program ICCC, Institut de Recerca de l’-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER-CV, Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Laura Casaní
- Cardiovascular Program ICCC, Institut de Recerca de l’-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Sandra Camino-Lopez
- Cardiovascular Program ICCC, Institut de Recerca de l’-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Oriol Juan-Babot
- Cardiovascular Program ICCC, Institut de Recerca de l’-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Institut de Recerca de l’-Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- CIBER-CV, Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
11
|
Singha SK, Muhammad I, Ibrahim MA, Wang M, Ashpole NM, Shariat-Madar Z. 4- O-Methylhonokiol Influences Normal Cardiovascular Development in Medaka Embryo. Molecules 2019; 24:molecules24030475. [PMID: 30699965 PMCID: PMC6384692 DOI: 10.3390/molecules24030475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/24/2019] [Accepted: 01/27/2019] [Indexed: 11/21/2022] Open
Abstract
Although 4-O-Methylhonokiol (MH) effects on neuronal and immune cells have been established, it is still unclear whether MH can cause a change in the structure and function of the cardiovascular system. The overarching goal of this study was to evaluate the effects of MH, isolated from Magnolia grandiflora, on the development of the heart and vasculature in a Japanese medaka model in vivo to predict human health risks. We analyzed the toxicity of MH in different life-stages of medaka embryos. MH uptake into medaka embryos was quantified. The LC50 of two different exposure windows (stages 9–36 (0–6 days post fertilization (dpf)) and 25–36 (2–6 dpf)) were 5.3 ± 0.1 μM and 9.9 ± 0.2 μM. Survival, deformities, days to hatch, and larval locomotor response were quantified. Wnt 1 was overexpressed in MH-treated embryos indicating deregulation of the Wnt signaling pathway, which was associated with spinal and cardiac ventricle deformities. Overexpression of major proinflammatory mediators and biomarkers of the heart were detected. Our results indicated that the differential sensitivity of MH in the embryos was developmental stage-specific. Furthermore, this study demonstrated that certain molecules can serve as promising markers at the transcriptional and phenotypical levels, responding to absorption of MH in the developing embryo.
Collapse
Affiliation(s)
- Santu K Singha
- Department of Biomolecular Sciences, Division of Pharmacology, University of Mississippi, University, MS 38677, USA.
| | - Ilias Muhammad
- The National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS 38677, USA.
| | - Mohamed Ali Ibrahim
- The National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS 38677, USA.
- Chemistry of Natural Compounds Department, National Research Centre, Dokki-Giza 12622, Egypt.
| | - Mei Wang
- The National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS 38677, USA.
| | - Nicole M Ashpole
- Department of Biomolecular Sciences, Division of Pharmacology, University of Mississippi, University, MS 38677, USA.
- The National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS 38677, USA.
| | - Zia Shariat-Madar
- Department of Biomolecular Sciences, Division of Pharmacology, University of Mississippi, University, MS 38677, USA.
- The National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, MS 38677, USA.
- Light Microscopy Core, University of Mississippi, University, MS 38677, USA.
| |
Collapse
|
12
|
Lietman C, Wu B, Lechner S, Shinar A, Sehgal M, Rossomacha E, Datta P, Sharma A, Gandhi R, Kapoor M, Young PP. Inhibition of Wnt/β-catenin signaling ameliorates osteoarthritis in a murine model of experimental osteoarthritis. JCI Insight 2018; 3:96308. [PMID: 29415892 PMCID: PMC5821202 DOI: 10.1172/jci.insight.96308] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/12/2017] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease involving both cartilage and synovium. The canonical Wnt/β-catenin pathway, which is activated in OA, is emerging as an important regulator of tissue repair and fibrosis. This study seeks to examine Wnt pathway effects on synovial fibroblasts and articular chondrocytes as well as the therapeutic effects of Wnt inhibition on OA disease severity. Mice underwent destabilization of the medial meniscus surgery and were treated by intra-articular injection with XAV-939, a small-molecule inhibitor of Wnt/β-catenin signaling. Wnt/β-catenin signaling was highly activated in murine synovial fibroblasts as well as in OA-derived human synovial fibroblasts. XAV-939 ameliorated OA severity associated with reduced cartilage degeneration and synovitis in vivo. Wnt inhibition using mechanistically distinct small-molecule inhibitors, XAV-939 and C113, attenuated the proliferation and type I collagen synthesis in synovial fibroblasts in vitro but did not affect human OA-derived chondrocyte proliferation. However, Wnt modulation increased COL2A1 and PRG4 transcripts, which are downregulated in chondrocytes in OA. In conclusion, therapeutic Wnt inhibition reduced disease severity in a model of traumatic OA via promoting anticatabolic effects on chondrocytes and antifibrotic effects on synovial fibroblasts and may be a promising class of drugs for the treatment of OA.
Collapse
Affiliation(s)
- Caressa Lietman
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Brian Wu
- Arthritis Program, University Health Network, Toronto, Ontario, Canada
| | - Sarah Lechner
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andrew Shinar
- Orthopedic Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Madhur Sehgal
- Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, and Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Evgeny Rossomacha
- Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, and Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Poulami Datta
- Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, and Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Anirudh Sharma
- Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, and Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Rajiv Gandhi
- Arthritis Program, University Health Network, Toronto, Ontario, Canada
| | - Mohit Kapoor
- Arthritis Program, University Health Network, Toronto, Ontario, Canada
- Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, and Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Pampee P. Young
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology and Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
13
|
Foulquier S, Daskalopoulos EP, Lluri G, Hermans KCM, Deb A, Blankesteijn WM. WNT Signaling in Cardiac and Vascular Disease. Pharmacol Rev 2018; 70:68-141. [PMID: 29247129 PMCID: PMC6040091 DOI: 10.1124/pr.117.013896] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
WNT signaling is an elaborate and complex collection of signal transduction pathways mediated by multiple signaling molecules. WNT signaling is critically important for developmental processes, including cell proliferation, differentiation and tissue patterning. Little WNT signaling activity is present in the cardiovascular system of healthy adults, but reactivation of the pathway is observed in many pathologies of heart and blood vessels. The high prevalence of these pathologies and their significant contribution to human disease burden has raised interest in WNT signaling as a potential target for therapeutic intervention. In this review, we first will focus on the constituents of the pathway and their regulation and the different signaling routes. Subsequently, the role of WNT signaling in cardiovascular development is addressed, followed by a detailed discussion of its involvement in vascular and cardiac disease. After highlighting the crosstalk between WNT, transforming growth factor-β and angiotensin II signaling, and the emerging role of WNT signaling in the regulation of stem cells, we provide an overview of drugs targeting the pathway at different levels. From the combined studies we conclude that, despite the sometimes conflicting experimental data, a general picture is emerging that excessive stimulation of WNT signaling adversely affects cardiovascular pathology. The rapidly increasing collection of drugs interfering at different levels of WNT signaling will allow the evaluation of therapeutic interventions in the pathway in relevant animal models of cardiovascular diseases and eventually in patients in the near future, translating the outcomes of the many preclinical studies into a clinically relevant context.
Collapse
Affiliation(s)
- Sébastien Foulquier
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Evangelos P Daskalopoulos
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Gentian Lluri
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Kevin C M Hermans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - Arjun Deb
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| | - W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands (S.F., K.C.M.H., W.M.B.); Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium (E.P.D.); Department of Medicine, Division of Cardiology, David Geffen School of Medicine (G.L., A.D.); and Department of Molecular Cell and Developmental Biology, University of California at Los Angeles, Los Angeles, California (A.D.)
| |
Collapse
|
14
|
Xu R, Bai Y, Zhao J, Xia H, Kong Y, Yao Z, Yan R, Zhang X, Hu X, Liu M, Yang Q, Luo G, Wu J. Silicone rubber membrane with specific pore size enhances wound regeneration. J Tissue Eng Regen Med 2017; 12:e905-e917. [DOI: 10.1002/term.2414] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/07/2017] [Accepted: 01/13/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Rui Xu
- Department of Neurology, Xinqiao Hospital & The Second Affiliated HospitalThird Military Medical University Chongqing China
| | - Yang Bai
- Institute of Burn Research, Southwest HospitalThird Military Medical University; State Key Laboratory of Trauma, Burn and Combined Injury; Chongqing Key Laboratory for Disease Proteomics Chongqing China
- Department of Otolaryngology, Southwest HospitalThird Military Medical University Chongqing China
| | - Jian Zhao
- State Key Laboratory of Polymer Materials EngineeringPolymer Research Institute of Sichuan University Chengdu China
| | - Hesheng Xia
- State Key Laboratory of Polymer Materials EngineeringPolymer Research Institute of Sichuan University Chengdu China
| | - Yi Kong
- Institute of Burn Research, Southwest HospitalThird Military Medical University; State Key Laboratory of Trauma, Burn and Combined Injury; Chongqing Key Laboratory for Disease Proteomics Chongqing China
| | - Zhihui Yao
- Institute of Burn Research, Southwest HospitalThird Military Medical University; State Key Laboratory of Trauma, Burn and Combined Injury; Chongqing Key Laboratory for Disease Proteomics Chongqing China
| | - Rongshuai Yan
- Institute of Burn Research, Southwest HospitalThird Military Medical University; State Key Laboratory of Trauma, Burn and Combined Injury; Chongqing Key Laboratory for Disease Proteomics Chongqing China
| | - Xiaorong Zhang
- Institute of Burn Research, Southwest HospitalThird Military Medical University; State Key Laboratory of Trauma, Burn and Combined Injury; Chongqing Key Laboratory for Disease Proteomics Chongqing China
| | - Xiaohong Hu
- Institute of Burn Research, Southwest HospitalThird Military Medical University; State Key Laboratory of Trauma, Burn and Combined Injury; Chongqing Key Laboratory for Disease Proteomics Chongqing China
| | - Meixi Liu
- Institute of Burn Research, Southwest HospitalThird Military Medical University; State Key Laboratory of Trauma, Burn and Combined Injury; Chongqing Key Laboratory for Disease Proteomics Chongqing China
| | - Qingwu Yang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated HospitalThird Military Medical University Chongqing China
| | - Gaoxing Luo
- Institute of Burn Research, Southwest HospitalThird Military Medical University; State Key Laboratory of Trauma, Burn and Combined Injury; Chongqing Key Laboratory for Disease Proteomics Chongqing China
| | - Jun Wu
- Institute of Burn Research, Southwest HospitalThird Military Medical University; State Key Laboratory of Trauma, Burn and Combined Injury; Chongqing Key Laboratory for Disease Proteomics Chongqing China
| |
Collapse
|
15
|
Gentzel M, Schambony A. Dishevelled Paralogs in Vertebrate Development: Redundant or Distinct? Front Cell Dev Biol 2017; 5:59. [PMID: 28603713 PMCID: PMC5445114 DOI: 10.3389/fcell.2017.00059] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/12/2017] [Indexed: 01/21/2023] Open
Abstract
Dishevelled (DVL) proteins are highly conserved in the animal kingdom and are important key players in β-Catenin-dependent and -independent Wnt signaling pathways. Vertebrate genomes typically comprise three DVL genes, DVL1, DVL2, and DVL3. Expression patterns and developmental functions of the three vertebrate DVL proteins however, are only partially redundant in any given species. Moreover, expression and function of DVL isoforms have diverged between different vertebrate species. All DVL proteins share basic functionality in Wnt signal transduction. Additional, paralog-specific interactions and functions combined with context-dependent availability of DVL isoforms may play a central role in defining Wnt signaling specificity and add selectivity toward distinct downstream pathways. In this review, we recapitulate briefly cellular functions of DVL paralogs, their role in vertebrate embryonic development and congenital disease.
Collapse
Affiliation(s)
- Marc Gentzel
- Molecular Analysis-Mass Spectrometry, Center for Molecular and Cellular Bioengineering (CMCB), TU DresdenDresden, Germany
| | - Alexandra Schambony
- Developmental Biology, Biology Department, Friedrich-Alexander University Erlangen-NurembergErlangen, Germany
| |
Collapse
|
16
|
Stylianidis V, Hermans KCM, Blankesteijn WM. Wnt Signaling in Cardiac Remodeling and Heart Failure. Handb Exp Pharmacol 2017; 243:371-393. [PMID: 27838851 DOI: 10.1007/164_2016_56] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Wnt signaling plays an essential role during development, but is also activated in diseases as diverse as neurodegeneration, osteoporosis, and cancer. Accumulating evidence demonstrates that Wnt signaling is also activated during cardiac remodeling and heart failure. In this chapter, we will provide a brief overview of Wnt signaling in all its complexity. Then we will discuss the evidence for its involvement in the development of cardiac hypertrophy, the wound healing after myocardial infarction (MI) and heart failure. Finally, we will provide an overview of the drugs that are available to target Wnt signaling at different levels of the signaling cascade and the results of these pharmacological interventions in cardiac disease.
Collapse
Affiliation(s)
- Vasili Stylianidis
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Kevin C M Hermans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|
17
|
Sun LY, Bie ZD, Zhang CH, Li H, Li LD, Yang J. MiR-154 directly suppresses DKK2 to activate Wnt signaling pathway and enhance activation of cardiac fibroblasts. Cell Biol Int 2016; 40:1271-1279. [PMID: 27542661 DOI: 10.1002/cbin.10655] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/14/2016] [Indexed: 12/26/2022]
Abstract
Excessive proliferation of cardiac fibroblasts (CFs) and their transdifferentiation into myofibroblasts leads to expression of α-smooth muscle actin (α-SMA), as well as excessive synthesis and secretion of collagens. This process represents an important pathological basis for myocardial fibrosis (MF). MicroRNA (miR)-154 and the Wnt signaling pathway play key roles in the above process, although their specific interactions are poorly understood. After transfecting CFs with miR-154 mimics or inhibitors, miR-154 was found to inhibit the expression of Dickkopf-related protein 2 (DKK2), while miR-154 inhibitors upregulated DKK2 expression in a Western blot analysis. In a subsequent dual-luciferase activity assay, direct binding of miR-154 to DKK2 was detected. Further experiments demonstrated that transfection of DKK2 siRNA or miR-154 resulted in increased levels of β-catenin, α-SMA, and collagens I and III. Moreover, these changes were observed in association with increases in CF proliferation and migration, and reduced apoptosis. Conversely, transfection of miR-154 inhibitors or DKK2 overexpression vector resulted in lower expression levels of β-catenin, α-SMA, and collagens I and III, suppressed cell proliferation and migration, and enhanced apoptosis. Furthermore, in each assay, when the DKK2 overexpression vector and miR-154 mimics were co-transfected, the functions of each component were counteracted by the other. Therefore, in CFs, targeting of DKK2 by miR-154 leads to upregulation of β-catenin expression and activation of the classical Wnt signaling pathway and CFs. These results suggest new targets for the clinical treatment of MF and ischemic heart disease.
Collapse
Affiliation(s)
- Li-Ye Sun
- Shandong University School of Medicine, Jinan, 250012, Shandong, China.,Department of Geratology, Yantai Yuhuangding Hospital, Yantai, 264000, Shandong, China
| | - Zi-Dong Bie
- Shandong University School of Medicine, Jinan, 250012, Shandong, China.,Department of Cardiology, Weihai Central Hospital, Weihai, 264423, Shandong, China
| | - Chuan-Huan Zhang
- Department of Cardiology, Yantai Yuhuangding Hospital, 20# Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
| | - Hua Li
- Department of Geratology, Yantai Yuhuangding Hospital, Yantai, 264000, Shandong, China
| | - Liu-Dong Li
- Department of Cardiology, Yantai Yuhuangding Hospital, 20# Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
| | - Jun Yang
- Department of Cardiology, Yantai Yuhuangding Hospital, 20# Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China
| |
Collapse
|
18
|
Abstract
Wnt signaling encompasses multiple and complex signaling cascades and is involved in many developmental processes such as tissue patterning, cell fate specification, and control of cell division. Consequently, accurate regulation of signaling activities is essential for proper embryonic development. Wnt signaling is mostly silent in the healthy adult organs but a reactivation of Wnt signaling is generally observed under pathological conditions. This has generated increasing interest in this pathway from a therapeutic point of view. In this review article, the involvement of Wnt signaling in cardiovascular development will be outlined, followed by its implication in myocardial infarct healing, cardiac hypertrophy, heart failure, arrhythmias, and atherosclerosis. The initial experiments not always offer consensus on the effects of activation or inactivation of the pathway, which may be attributed to (i) the type of cardiac disease, (ii) timing of the intervention, and (iii) type of cells that are targeted. Therefore, more research is needed to determine the exact implication of Wnt signaling in the conditions mentioned above to exploit it as a powerful therapeutic target.
Collapse
|
19
|
Matthijs Blankesteijn W, Hermans KCM. Wnt signaling in atherosclerosis. Eur J Pharmacol 2015; 763:122-30. [PMID: 25987418 DOI: 10.1016/j.ejphar.2015.05.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/01/2015] [Indexed: 02/03/2023]
Abstract
Atherosclerosis is a disease of the vascular wall that forms the basis for a large spectrum of pathologies of various organs and tissues. Although massive research efforts in the last decades have yielded valuable information about its underlying molecular mechanisms, this has not led to a translation into effective therapeutic interventions that can stop the progression or even can induce regression of atherosclerosis. This underscores the importance of investigations on the involvement of novel signaling pathways in the development and progression of this condition. In this review we focus on the role of Wnt signaling in atherosclerosis. Experimental evidence is presented that Wnt signaling is involved in many aspects of the development and progression of vascular lesions including endothelial dysfunction, macrophage activation and the proliferation and migration of vascular smooth muscle cells. Subsequently, we will discuss the role of Wnt signaling in myocardial infarction and stroke, two common pathologies resulting from the progression of atherosclerotic lesions towards an unstable phenotype. Despite the fact that the published data sometimes are ambiguous or even conflicting, a picture is emerging that an attenuation of Wnt signaling is beneficial for the cardiovascular system that is compromised by atherosclerosis.
Collapse
Affiliation(s)
- W Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht, Maastricht University, P.O. Box 616, 6200MD Maastricht, The Netherlands.
| | - Kevin C M Hermans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht, Maastricht University, P.O. Box 616, 6200MD Maastricht, The Netherlands
| |
Collapse
|
20
|
Roche PL, Filomeno KL, Bagchi RA, Czubryt MP. Intracellular Signaling of Cardiac Fibroblasts. Compr Physiol 2015; 5:721-60. [DOI: 10.1002/cphy.c140044] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
21
|
Hua J, Xu Y, He Y, Jiang X, Ye W, Pan Z. Wnt4/β-catenin signaling pathway modulates balloon-injured carotid artery restenosis via disheveled-1. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:8421-8431. [PMID: 25674206 PMCID: PMC4314008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 11/26/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND Restenosis is a common adverse event of endovascular procedures and troubles cardiologists. However, the mechanism underlying restenosis is still not fully understood. To evaluate whether disheveled-1 (Dvl-1) is involved in the Wnt4/β-catenin signaling pathway to participate in the mechanisms of vascular restenosis. METHODOLOGY Rat model of balloon-injured carotid artery was established and atorvastatin was used to treat artery injury. Vascular smooth muscle cells (VSMC) were isolated from rats and cultured in DMEM exposed to AngII. Down-regulation and overexpression of Dvl-1 were conducted in cells to explore the role underlying its effects on VSMC proliferation and collagen expression. Adenovirus with overexpressing Dvl-1 was injected into rats to evaluate the role of Dvl-1 in artery injury rats. RESULTS The results in vivo found that Wnt4, Dvl-1 and β-catenin expression as well as collagen volume fraction (CVF) in injured artery were significantly increased. The results in vitro showed that Dvl-1 overexpression reversed the treatment effects of atorvastatin on VSMCs proliferation and collagen expression. It was also canceled by overexpressing Dvl-1 that the decrease of β-catenin protein treated with atorvastatin in cells exposed to AngII. In addition, treated artery injury rats with atorvastatin, the group with injection of Ad-Dvl-1 had higher levels of intima thickness, intimal/medial area ratio and CVF. CONCLUSION Dvl-1 was probably a key regulator in the pathway of wnt4/β-catenin to take part in the vascular restenosis partly, and Dvl-1 is a potential gene to anti- restenosis.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Angioplasty, Balloon/adverse effects
- Animals
- Blotting, Western
- Carotid Artery Injuries/metabolism
- Cells, Cultured
- Disease Models, Animal
- Dishevelled Proteins
- Graft Occlusion, Vascular/metabolism
- Immunohistochemistry
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Phosphoproteins/metabolism
- Rats
- Rats, Sprague-Dawley
- Real-Time Polymerase Chain Reaction
- Wnt Signaling Pathway/physiology
- Wnt4 Protein/metabolism
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Junyi Hua
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University of Traditional Chinese Medicine Hangzhou 310006, Zhengjiang, China
| | - Yun Xu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University of Traditional Chinese Medicine Hangzhou 310006, Zhengjiang, China
| | - Yuzhou He
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University of Traditional Chinese Medicine Hangzhou 310006, Zhengjiang, China
| | - Xuhong Jiang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University of Traditional Chinese Medicine Hangzhou 310006, Zhengjiang, China
| | - Wu Ye
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University of Traditional Chinese Medicine Hangzhou 310006, Zhengjiang, China
| | - Zhimin Pan
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University of Traditional Chinese Medicine Hangzhou 310006, Zhengjiang, China
| |
Collapse
|
22
|
|
23
|
Borrell-Pagès M, Romero JC, Badimon L. LRP5 negatively regulates differentiation of monocytes through abrogation of Wnt signalling. J Cell Mol Med 2013; 18:314-25. [PMID: 24266894 PMCID: PMC3930418 DOI: 10.1111/jcmm.12190] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/17/2013] [Indexed: 01/22/2023] Open
Abstract
Molecular changes involved in cell differentiation are only partially known. Circulating inflammatory cells need to differentiate to perform specialized functions in target tissues. Here, we hypothesized that low-density lipoprotein receptor–related protein 5 (LRP5) is involved, through its participation in the canonical Wnt/β-catenin signalling, in the differentiation process of monocytic cells. To this aim, we characterized differentiation mechanisms of HL60 cells and primary human monocytes. We show that silencing the LRP5 gene increased differentiation of HL60 cells and human monocytes, suggesting that LRP5 signalling abrogates differentiation. We demonstrate that the mechanisms behind this blockade include sequestration of β-catenin at the cellular membrane, inhibition of the Wnt signalling and increase of apoptosis. We further demonstrate the involvement of LRP5 and the Wnt/β-catenin signalling in the process because cellular differentiation can be rescued by the addition of downstream Wnt target genes to the monocytic cells.
Collapse
Affiliation(s)
- Maria Borrell-Pagès
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | | | | |
Collapse
|
24
|
Abstract
Myocardial infarction is one of the major causes of left ventricular dilatation, frequently leading to heart failure. In the last decade, the wound healing process that takes place in the infarct area after infarction has been recognized as a novel therapeutic target to attenuate left ventricular dilatation and preserve an adequate cardiac function. In this chapter, we discuss the role of Wnt signaling in the wound healing process after infarction, with a specific focus on its modulating effect on myofibroblast characteristics.
Collapse
|
25
|
Dawson K, Aflaki M, Nattel S. Role of the Wnt-Frizzled system in cardiac pathophysiology: a rapidly developing, poorly understood area with enormous potential. J Physiol 2012. [PMID: 23207593 DOI: 10.1113/jphysiol.2012.235382] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract The Wnt-Frizzled (Fzd) G-protein-coupled receptor system, involving 19 distinct Wnt ligands and 10 Fzd receptors, plays key roles in the development and functioning of many organ systems. There is increasing evidence that Wnt-Fzd signalling is important in regulating cardiac function. Wnt-Fzd signalling primarily involves a canonical pathway, with dishevelled-1-dependent nuclear translocation of β-catenin that derepresses Wnt-sensitive gene transcription, but can also include non-canonical pathways via phospholipase-C/Ca(2+) mobilization and dishevelled-protein activation of small GTPases. Wnt-Fzd effects vary with specific ligand/receptor interactions and associated downstream pathways. This paper reviews the biochemistry and physiology of the Wnt-Fzd complex, and presents current knowledge of Wnt signalling in cardiac remodelling processes such as hypertrophy and fibrosis, as well as disease states such as myocardial infarction (MI), heart failure and arrhythmias. Wnt signalling is activated during hypertrophy; inhibiting Wnt signalling by activating glycogen synthase kinase attenuates the hypertrophic response. Wnt signalling has complex and time-dependent actions post-MI, so that either beneficial or harmful effects might result from Wnt-directed interventions. Stem cell biology, a promising area for therapeutic intervention, is highly regulated by Wnt signalling. The Wnt system regulates fibroblast function, and is prominently altered in arrhythmogenic ventricular cardiomyopathy, a familial disease involving excess deposition of fibroadipose tissue. Wnt signalling controls connexin43 expression, thereby contributing to the regulation of cardiac electrical stability and arrhythmia generation. Although much has been learned about Wnt-Fzd signalling in hypertrophy and infarction, its role is poorly understood for a broad range of other heart disorders. Much more needs to be learned for its contributions to be fully appreciated, and to permit more effective exploitation of its enormous potential in therapeutic development.
Collapse
Affiliation(s)
- Kristin Dawson
- S. Nattel: 5000 Belanger St. E, Montreal, Quebec, Canada H1T 1C8.
| | | | | |
Collapse
|
26
|
Hermans KC, Daskalopoulos EP, Blankesteijn WM. Interventions in Wnt signaling as a novel therapeutic approach to improve myocardial infarct healing. FIBROGENESIS & TISSUE REPAIR 2012; 5:16. [PMID: 22967504 PMCID: PMC3472244 DOI: 10.1186/1755-1536-5-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 08/20/2012] [Indexed: 01/12/2023]
Abstract
Following myocardial infarction, wound healing takes place in the infarct area where the non-viable cardiac tissue is replaced by a scar. Inadequate wound healing or insufficient maintenance of the extracellular matrix in the scar can lead to excessive dilatation of the ventricles, one of the hallmarks of congestive heart failure. Therefore, it is important to better understand the wound-healing process in the heart and to develop new therapeutic agents that target the infarct area in order to maintain an adequate cardiac function. One of these potential novel therapeutic targets is Wnt signaling. Wnt signaling plays an important role in embryonic myocardial development but in the adult heart the pathway is thought to be silent. However, there is increasing evidence that components of the Wnt pathway are re-expressed during cardiac repair, implying a regulatory role. Recently, several studies have been published where the effect of interventions in Wnt signaling on infarct healing has been studied. In this review, we will summarize the results of these studies and discuss the effects of these interventions on the different cell types that are involved in the wound healing process.
Collapse
Affiliation(s)
- Kevin Cm Hermans
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Maastricht University, 50 Universiteitssingel, 6229ER Maastricht, PO Box 616 6200MD, Maastricht, The Netherlands.
| | | | | |
Collapse
|
27
|
Abstract
Wnt signaling is activated by wounding and participates in every subsequent stage of the healing process from the control of inflammation and programmed cell death, to the mobilization of stem cell reservoirs within the wound site. In this review we summarize recent data elucidating the roles that the Wnt pathway plays in the injury repair process. These data provide a foundation for potential Wnt-based therapeutic strategies aimed at stimulating tissue regeneration.
Collapse
Affiliation(s)
- Jemima L Whyte
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California 94305, USA
| | | | | |
Collapse
|
28
|
Abstract
Emerging data in the field of cardiac development as well as repair and regeneration indicate a complex and important interplay between endocardial, epicardial, and myofibroblast populations that is critical for cardiomyocyte differentiation and postnatal function. For example, epicardial cells have been shown to generate cardiac myofibroblasts and may be one of the primary sources for this cell lineage during development. Moreover, paracrine signaling from the epicardium and endocardium is critical for proper development of the heart and pathways such as Wnt, fibroblast growth factor, and retinoic acid signaling have been shown to be key players in this process. Despite this progress, interactions between nonmyocyte cells and cardiomyocytes in the heart are still poorly understood. We review the various nonmyocyte-myocyte interactions that occur in the heart and how these interactions, primarily through signaling networks, help direct cardiomyocyte differentiation and regulate postnatal cardiac function.
Collapse
Affiliation(s)
- Ying Tian
- Department of Medicine, University of Pennsylvania, PA 19104-5129, USA
| | | |
Collapse
|
29
|
Exogenous high-mobility group box 1 protein injection improves cardiac function after myocardial infarction: involvement of Wnt signaling activation. J Biomed Biotechnol 2012; 2012:743879. [PMID: 22675257 PMCID: PMC3364756 DOI: 10.1155/2012/743879] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 03/20/2012] [Accepted: 03/22/2012] [Indexed: 12/31/2022] Open
Abstract
Exogenous high-mobility group box 1 protein (HMGB1) injection could prevent left ventricular remodeling and enhance left ventricular function during myocardial infarction (MI). However, the mechanism remains unclear. This paper was to investigate in the mechanism of cardioprotection of HMGB1 during MI in rats. Anesthetized male rats were treated once with HMGB1 (200 ng) 4 h after MI and then executed after 7 and 28 days, respectively. Cardiac function, collagen deposition, and dishevelled-1 and β-catenin protein expression were measured. After MI 7 days or 28 days, the left ventricular ejection fraction (LVEF) was significantly decreased compared to that of sham-operated control group (P < 0.05). However, the LVEF HMGB1-treated groups were significantly higher compared to those of the MI group in both 7 days and 28 days (P < 0.05). The collagen volume fraction was significantly reduced in the HMGB1-treated group in infarcted border zone. HMGB1 could activate the expression of dishevelled-1 and β-catenin proteins (P < 0.05). Our study suggested that exogenous high-mobility group box 1 protein injection improves cardiac function after MI, which may be involved in Wnt/β-catenin signaling activation.
Collapse
|
30
|
Aisagbonhi O, Rai M, Ryzhov S, Atria N, Feoktistov I, Hatzopoulos AK. Experimental myocardial infarction triggers canonical Wnt signaling and endothelial-to-mesenchymal transition. Dis Model Mech 2011; 4:469-83. [PMID: 21324930 PMCID: PMC3124051 DOI: 10.1242/dmm.006510] [Citation(s) in RCA: 223] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite available therapies, myocardial infarction (MI) remains a leading cause of death worldwide. Better understanding of the molecular and cellular mechanisms that regulate cardiac repair should help to improve the clinical outcome of MI patients. Using the reporter mouse line TOPGAL, we show that canonical (β-catenin-dependent) Wnt signaling is induced 4 days after experimental MI in subepicardial endothelial cells and perivascular smooth muscle actin (SMA)-positive (SMA+) cells. At 1 week after ischemic injury, a large number of canonical-Wnt-positive cells accumulated in the infarct area during granulation tissue formation. Coincidently with canonical Wnt activation, endothelial-to-mesenchymal transition (EndMT) was also triggered after MI. Using cell lineage tracing, we show that a significant portion of the canonical-Wnt-marked SMA+ mesenchymal cells is derived from endothelial cells. Canonical Wnt signaling induces mesenchymal characteristics in cultured endothelial cells, suggesting a direct role in EndMT. In conclusion, our study demonstrates that canonical Wnt activation and EndMT are molecular and cellular responses to MI and that canonical Wnt signaling activity is a characteristic property of EndMT-derived mesenchymal cells that take part in cardiac tissue repair after MI. These findings could lead to new strategies to improve the course of cardiac repair by temporal and cell-type-specific manipulation of canonical Wnt signaling.
Collapse
Affiliation(s)
- Omonigho Aisagbonhi
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University, 2213 Garland Avenue, Nashville, TN 37232-6300, USA
| | | | | | | | | | | |
Collapse
|
31
|
Oerlemans MIFJ, Goumans MJ, van Middelaar B, Clevers H, Doevendans PA, Sluijter JPG. Active Wnt signaling in response to cardiac injury. Basic Res Cardiol 2010; 105:631-41. [PMID: 20373104 PMCID: PMC2916122 DOI: 10.1007/s00395-010-0100-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 03/16/2010] [Accepted: 03/23/2010] [Indexed: 12/14/2022]
Abstract
Although the contribution of Wnt signaling in infarct healing is suggested, its exact role after myocardial infarction (MI) still needs to be unraveled. We evaluated the cardiac presence of active Wnt signaling in vivo following MI, and investigated in which cell types active Wnt signaling was present by determining Axin2 promoter-driven LacZ expression. C57BL/6 Axin2-LacZ reporter mice were sacrificed at days 0, 1, 3, 7, 14, and 21 after LAD ligation. Hearts were snap-frozen for immunohistochemistry (IHC) or enzymatically digested to obtain a single cell suspension for flow cytometric analysis. For both FACS and IHC, samples were stained for β-galactosidase and antibodies against Sca-1, CD31, ckit, and CD45. Active Wnt signaling increased markedly in the myocardium, from 7 days post-MI onwards. Using Sca-1 and CD31, to identify progenitor and endothelial cells, a significant increase in LacZ+ cells was found at 7 and 14 days post-MI. LacZ+ cells also increased in the ckit+ and CD45+ cell population. IHC revealed LacZ+ cells co-expressing Sca, CD31, CD45, vWF, and αSMA in the border zone and the infarcted area. Wnt signaling increased significantly after MI in Sca+- and CD31+-expressing cells, suggesting involvement of Wnt signaling in resident Sca+ progenitor cells, as well as endothelial cells. Moreover, active Wnt signaling was present in ckit+ cells, leukocytes, and fibroblast. Given its broad role during the healing phase after cardiac injury, additional research seems warranted before a therapeutic approach on Wnt to enhance cardiac regeneration can be carried out safely.
Collapse
|
32
|
Abstract
Cardiac development is comprised of a series of morphological events tightly controlled both spatially and temporally. The molecular pathways controlling early cardiac differentiation are poorly understood, but Wnt signaling is emerging as a critical pathway for multiple aspects of early cardiovascular development. The Wnt pathway plays multiple roles in regulating cellular behavior including proliferation, differentiation, cell migration, and cell polarity. Recent data have demonstrated that Wnt activity is important for early precardiac mesoderm differentiation but must be inhibited in subsequent steps for cardiomyocyte differentiation to proceed. Given the important role that Wnt signaling plays in both the differentiation of cardiomyocytes from pluripotential stem cells and tissue regeneration in general, an increased understanding of this pathway is likely to enhance our knowledge about both cardiovascular development and reparative mechanisms.
Collapse
|
33
|
Laeremans H, Rensen SS, Ottenheijm HCJ, Smits JFM, Blankesteijn WM. Wnt/frizzled signalling modulates the migration and differentiation of immortalized cardiac fibroblasts. Cardiovasc Res 2010; 87:514-23. [PMID: 20189955 DOI: 10.1093/cvr/cvq067] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS The Wnt/frizzled (Fzd) signal transduction cascade has been implicated in the proliferation, differentiation, and migration of many cell types, but the role of this pathway in cardiac fibroblast differentiation is not known. Our lab previously showed an up-regulation of Fzd-1 and -2 expression in myofibroblasts after myocardial infarction (MI), indicating a potential role for the Fzd receptor in fibroblast-myofibroblast differentiation. The present study was performed to further define the role of specific Wnt and Fzd proteins in the proliferation, migration, and differentiation of cardiac fibroblasts. METHODS AND RESULTS Because primary fibroblasts become senescent after a few passages and are difficult to transfect, we immortalized rat cardiac fibroblasts with telomerase [cardiac fibroblasts immortalized with telomerase (CFIT)]. Proliferation of CFIT was not significantly influenced by Wnt/Fzd signalling. The migration, however, was attenuated by all Wnt/Fzd combinations tested. Also, specific Wnt/Fzd combinations modulated the expression of the following myofibroblast markers: collagen Ialpha1, collagen III, fibronectin and its splice variants, and alpha-smooth muscle actin. CONCLUSION The results indicate that myofibroblast migration and differentiation, but not proliferation, can be modulated by interventions in Wnt/Fzd signalling. Therefore, Wnt/Fzd signalling may serve as a novel therapeutic target to ameliorate wound healing after MI.
Collapse
Affiliation(s)
- Hilde Laeremans
- Department of Pharmacology and Toxicology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 50 Universiteitssingel, PO Box 616, 6200 MD Maastricht, The Netherlands
| | | | | | | | | |
Collapse
|
34
|
Xiong M, Qu J, Zhou LP, Zhang DJ, Qiao YY, Hu M, Zhao XH. Seawater immersion depresses the migration of vascular endothelial cell and inhibits angiogenesis. Shijie Huaren Xiaohua Zazhi 2009; 17:2152-2158. [DOI: 10.11569/wcjd.v17.i21.2152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of seawater immersion on vascular endothelial cell migration and angiogenesis.
METHODS: A scratch wound model was established using ECV304 cells. Cells were divided into control group (immersed in normal saline) and experimental group (immersed in seawater). Cell migration was examined using the scratch wound healing recovery assay. A nude mouse xenograft model was developed by subcutaneous inoculation of esophageal squamous cells (ECO156) in nude mice. Tumors were also divided into control group (immersed in normal saline) and experimental group (immersed in seawater). Tumor volume and weight were measured, and tumor growth curves were plotted. Neovascular vessels surrounding the tumor were observed, and microvessel density was detected by immunohistochemical staining.
RESULTS: After seawater immersion, ECV304 cells lost their normal morphology, and showed obvious dehydration with the prolongation of immersion duration. Seventeen hours after wounding, the scratch wound in the control group had almost completely healed, while that in the experiment group was still obvious, suggesting that the healing of scratch wound in the experiment group was delayed significantly and the migration of cells was depressed. The microvessel density in the experiment group was significantly lower than that in the control group (1.20 ± 0.44 vs 3.20 ± 0.83, P < 0.05). Compared with the control group, tumors in the experiment group grew slowly and had significantly smaller size (P < 0.05).
CONCLUSION: Seawater immersion may depress the migration of vascular endothelial cells and inhibit angiogenesis.
Collapse
|
35
|
Cohen ED, Tian Y, Morrisey EE. Wnt signaling: an essential regulator of cardiovascular differentiation, morphogenesis and progenitor self-renewal. Development 2008; 135:789-98. [PMID: 18263841 DOI: 10.1242/dev.016865] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Emerging evidence indicates that Wnt signaling regulates crucial aspects of cardiovascular biology (including cardiac morphogenesis, and the self-renewal and differentiation of cardiac progenitor cells). The ability of Wnt signaling to regulate such diverse aspects of cardiovascular development rests on the multifarious downstream and tangential targets affected by this pathway. Here, we discuss the roles for Wnt signaling in cardiac and vascular development, and focus on the emerging role of Wnt signaling in cardiovascular morphogenesis and progenitor cell self-renewal.
Collapse
Affiliation(s)
- Ethan David Cohen
- Cardiovascular Institute, Institute for Regenerative Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
36
|
van de Schans VA, Smits JF, Blankesteijn WM. The Wnt/frizzled pathway in cardiovascular development and disease: Friend or foe? Eur J Pharmacol 2008; 585:338-45. [DOI: 10.1016/j.ejphar.2008.02.093] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2007] [Revised: 01/25/2008] [Accepted: 02/06/2008] [Indexed: 01/19/2023]
|
37
|
Maiese K, Li F, Chong ZZ, Shang YC. The Wnt signaling pathway: aging gracefully as a protectionist? Pharmacol Ther 2008; 118:58-81. [PMID: 18313758 DOI: 10.1016/j.pharmthera.2008.01.004] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Accepted: 01/18/2008] [Indexed: 12/16/2022]
Abstract
No longer considered to be exclusive to cellular developmental pathways, the Wnt family of secreted cysteine-rich glycosylated proteins has emerged as versatile targets for a variety of conditions that involve cardiovascular disease, aging, cancer, diabetes, neurodegeneration, and inflammation. In particular, modulation of Wnt signaling may fill a critical void for the treatment of disorders that impact upon both cellular survival and cellular longevity. Yet, in some scenarios, Wnt signaling can become the catalyst for disease development or promote cell senescence that can compromise clinical utility. This double edge sword in regards to the role of Wnt and its signaling pathways highlights the critical need to further elucidate the cellular mechanisms governed by Wnt in conjunction with the development of robust pharmacological ligands that may open new avenues for disease treatment. Here we discuss the influence of the Wnt pathway during cell survival, metabolism, and aging in order for one to gain a greater insight for the novel role of Wnt signaling as well as exemplify its unique cellular pathways that influence both normal physiology and disease.
Collapse
Affiliation(s)
- Kenneth Maiese
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201, USA.
| | | | | | | |
Collapse
|
38
|
Li F, Chong ZZ, Maiese K. Winding through the WNT pathway during cellular development and demise. Histol Histopathol 2006; 21:103-24. [PMID: 16267791 PMCID: PMC2247407 DOI: 10.14670/hh-21.103] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In slightly over a period of twenty years, our comprehension of the cellular and molecular mechanisms that govern the Wnt signaling pathway continue to unfold. The Wnt proteins were initially implicated in viral carcinogenesis experiments associated with mammary tumors, but since this period investigations focusing on the Wnt pathways and their transmembrane receptors termed Frizzled have been advanced to demonstrate the critical nature of Wnt for the development of a variety of cell populations as well as the potential of the Wnt pathway to avert apoptotic injury. In particular, Wnt signaling plays a significant role in both the cardiovascular and nervous systems during embryonic cell patterning, proliferation, differentiation, and orientation. Furthermore, modulation of Wnt signaling under specific cellular influences can either promote or prevent the early and late stages of apoptotic cellular injury in neurons, endothelial cells, vascular smooth muscle cells, and cardiomyocytes. A number of downstream signal transduction pathways can mediate the biological response of the Wnt proteins that include Dishevelled, beta-catenin, intracellular calcium, protein kinase C, Akt, and glycogen synthase kinase-3beta. Interestingly, these cellular cascades of the Wnt-Frizzled pathways can participate in several neurodegenerative, vascular, and cardiac disorders and may be closely integrated with the function of trophic factors. Identification of the critical elements that modulate the Wnt-Frizzled signaling pathway should continue to unlock the potential of Wnt pathway for the development of new therapeutic options against neurodegenerative and vascular diseases.
Collapse
Affiliation(s)
- F Li
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
39
|
Masaki M, Izumi M, Oshima Y, Nakaoka Y, Kuroda T, Kimura R, Sugiyama S, Terai K, Kitakaze M, Yamauchi-Takihara K, Kawase I, Hirota H. Smad1 Protects Cardiomyocytes From Ischemia-Reperfusion Injury. Circulation 2005; 111:2752-9. [PMID: 15911698 DOI: 10.1161/circulationaha.104.490946] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
We previously reported that bone morphogenetic protein 2 (BMP2) protected against apoptosis of serum-deprived cardiomyocytes via induction of Bcl-xL through the Smad1 pathway. To investigate whether Smad1 signaling promotes cell survival in the adult heart, we subjected transgenic mice with cardiac-specific overexpression of smad1 gene (Smad1TG) to ischemia-reperfusion (I/R) injury.
Methods and Results—
The effects of BMP2 or adenovirus-mediated transfection of smad1 on cardiomyocyte survival in hypoxia-reoxygenation were examined using rat neonatal cardiomyocytes. BMP2 and Smad1 each significantly promoted survival and diminished apoptotic death of cardiomyocytes during hypoxia-reoxygenation. Interestingly, Smad1 was found to be activated during I/R in normal mouse heart. To examine physiological and pathological roles of Smad1 in I/R, we generated Smad1TG using the α-myosin heavy chain gene promoter. Phosphorylation of Smad1 was found in all smad1 transgene–positive mouse hearts. To examine whether Smad1 prevents injury of cardiomyocytes in vivo, we subjected Smad1TG and age-matched wild-type mice (WT) to I/R injury induced by 1 hour of ligation of the left coronary artery and 1 hour of reperfusion. TUNEL and DNA ladder analyses showed that Smad1TG had significantly smaller myocardial infarctions and fewer apoptotic deaths of cardiomyocytes than did WT. Interestingly, increased expression of Bcl-xL and β-catenin was more remarkable whereas caspase3 was less activated in Smad1TG heart than in that of WT.
Conclusions—
These findings suggest that the Smad1 signaling pathway plays a role in cardioprotection against I/R injury.
Collapse
Affiliation(s)
- Mitsuru Masaki
- Department of Molecular Medicine, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita City, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|